Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 ISSN 0976 – 044X Research Article Updates on the Biochemical and Molecular Mechanisms of N-nitrosodiethylamine-induced Hepatocellular Carcinoma: Promising Therapeutic Role of Punica granatum Peel Extract *1 2 2 3 2 Hanaa H. Ahmed , Wafaa Gh. Shousha , Hatem A. El-Mezayen , Sayed A. El-Toumy , Aesha R. Ramadan 1 Hormones Department, National Research Centre, Dokki, Giza, Egypt. 2 Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt. 3 Chemistry of Tannins Department, National Research Centre, Dokki, Giza, Egypt. *Corresponding author’s E-mail: hanaaomr@yahoo.com Accepted on: 14-04-2015; Finalized on: 31-05-2015. ABSTRACT This study was undertaken to elucidate the underlying biochemical and molecular mechanisms in favour of N-nitrosodiethylamineinduced hepatocellular carcinoma. Also, the aim of this work was extended to explore the efficacy of Punica granatum peel extract in retrogression of hepatocellular carcinoma (HCC) in rats. HCC group experienced significant elevation in the studied biochemical markers in serum paralleled by significant downregulation of ING-3 gene expression level and upregulation of Foxp-1 gene expression level in liver. Moreover, HCC group exhibited remarkable increase in β-catenin, survivin and Ki-67 expression in the liver as shown in the immunohistochemical analysis. Furthermore, histopathological investigation of liver tissue sections of rats in HCC group revealed typical anaplasia. In contrast, the treatment of HCC groups with Punica granatum peel extract resulted in significant depletion in the liver enzymes activity in association with significant reduction in the studied tumor markers and angiogenic markers. Upregulation of ING-3 and downregulation of Foxp-1 gene expression level in liver have been also detected due to treatment of HCC groups with Punica granatum peel extract. Furthermore, these groups displayed marked decrease in β-catenin, survivin and Ki-67 expression in the liver. Interestingly, treatment with Punica granatum peel extract elicited marked improvement in the histological feature of liver tissue of HCC groups. In conclusion, the present study indicated that the carcinogenic potency of N-nitrosodiethylamine targeted multiple systems on the cellular and molecular levels. Also, the present findings shed light on the promising anticancer activity of Punica granatum peel extract in recession of hepatocellular carcinoma induced chemically in the experimental model through its antiangiogenic, apoptotic and antiproliferative effciency. Keywords: Hepatocellular carcinoma, Punica granatum, Tumor markers, Angiogenesis, Apoptosis, Proliferation. INTRODUCTION H epatocellular carcinoma (HCC), like any other cancer, develops when there is a mutation to the cellular machinery that causes the cell to replicate at a higher rate and results in the cell growth avoiding apoptosis.1 Among primary liver cancers, HCC represents the major histological subtype, accounting for 70% to 85% of the total liver cancer burden worldwide.2 Globally, it is the fifth most common cancer and the second leading cause of cancer-related death.3 In developing countries, incidence rates of HCC are two to three fold higher than in developed countries.4 In Egypt, HCC contributes to 14.8% of all cancer mortality with a higher incidence in males (17.3%) than in females (11.5%). It is the second most frequent cancer type in Egyptian males after bladder cancer and the eighth most 5 frequent in Egyptian females. Hospital-based studies have reported an overall increase in the relative frequency of all liver-related cancers in Egypt.6 This rising incidence may be due to high prevalence of hepatitis C virus (HCV) and its complications and the fact that people born 20 years ago or earlier in Egypt has not been vaccinated against hepatitis B virus (HBV).7,8 Chronic inflammations of the liver and subsequent cirrhosis are highly correlated with hepatitis B and hepatitis C viral infections, alcohol abuse and metabolic liver diseases (diabetes and non alcoholic fatty liver disease) these are considered as the major risk factors for HCC. Additionally, obesity, environmental pollutants, consumption of food contaminated with the fungal toxins as aflatoxins that are produced by Aspergillus flavus in food grains and nitrosamine consumption are the strongest risk factors for HCC development.9 N-nitrosodiethylamine (NDEA) is an N-nitroso alkyl compound described as an effective hepatotoxin in the experimental animals, producing toxicity after repeated administration.10 NDEA is found in a wide variety of foods such as cheese, soybeans, smoked, salted and dried fish, 11 cured meat and alcoholic beverages. NDEA is mainly metabolized in the liver by the action of cytochrome p450 enzymes and its reactive metabolites are primarily responsible for its hepatotoxic effects. NDEA is bioactivated to ethyldiazonium ion which alkylates DNA bases to form pro-mutagenic adducts such as O6ethyldeoxyguanosine and O4 and O6ethyldeoxythymidine and these bioactive molecules induce oxidative stress and cytotoxicity by damaging biomolecules such as DNA, lipids and proteins.12 Natural products are non-toxic natural extracts or compounds that compared with synthetic materials, generally produce less side effects.13 Thus, they potentially provide an alternative or adjunct therapeutic International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 121 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 option for patients with cancer. According to the holy Quran, pomegranates grow in the gardens of paradise and the Quran has recited the Punica granutum twice as 14 an example of god’s good creations. Punica granutum, popularly known as pomegranate, is belonging to Punicaceae family. The extract of Punica granatum has been reported to have various medicinal values; antioxidant, antibacterial, antidiabetic, cardioprotective and anticarcinogenic activity.15-19 Punica granatum peel is a rich source of antioxidants, specially polyphenols, such as ellagic acid, querectin, kaempferol, luteolin and punicalagin.20,21 The current study was designed to elucidate the biochemical and molecular mechanisms of Nnitrosodiethylamine-induced hepatocellular carcinomain rats. Also, the present goal was extended to explore the potency of Punica granatum peel extract in retraction of hepatocellular carcinoma with special concern on its mechanism of action. MATERIALS AND METHODS Materials Chemicals and Drug N-nitrosodiethylamine (NDEA) (CAS no. 55-18-5) was purchased from Sigma-Aldrich Chemicals Co. (St Louis, MO, USA). Doxorubicin was supplied from Pharmacia Italia S.P.A. Milan, Italy. All other chemicals used in the present study were of high analytical grade. Plant Materials Punica granatum fruits were purchased from local market, Cairo, Egypt. The plant was authenticated by Professor Ibrahim El-Garf, Professor of Plant Taxonomy, Department of Botany, Faculty of Science, Cairo University. Voucher Speciemens were kept in the museum of the Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, Egypt. Preparation of Punica granatum Peel Extract Three kilograms of Punica granatum peel were separated from seeds of Punica granatum fruits. The peels were cut into small pieces and blended with 3000 ml of methanol 70% using electric blender. Then, the mixture was left for 72 hrs and filtered using filter paper. The filtrate was evaporated under reduced pressure at 45-50 ˚C using rotary evaporator to give a dry residue of crude aqueous methanolic extract (16.5% from fresh peel).22 Animals and Experimental Protocol A total number of 60 adult female albino rats of Wister strain weighing 120-150 g were used in the present study. The animals were obtained from the Animal House Colony of the National Research Centre, Giza, Egypt. The animals were housed in polypropylene cages in an environmentally controlled clean air room with a temperature of 25±1˚C, an alternating 12h light/12h dark cycle, a relative humidity of 60 ± 5% and free access to ISSN 0976 – 044X tap water and a standard rodent chow (Wadi El Kabda Co., Cairo, Egypt). The animals were allowed to adapt to these conditions for 2 weeks before the beginning of the experimental protocol. The animal experimental protocol was approved by the Ethical Committee for Medical Research, National Research Centre, Egypt. After the acclimatization period, the animals were divided into 5 groups (12 rats/group) as follows: 1) Normal healthy rats received 1ml/rat normal saline orally and served as negative control group. 2) Hepatocellular carcinoma (HCC) group in which the rats were orally administered with NDEA (dissolved in 0.9% normal saline), in a dose of 20 mg/kg b.wt. five times a week during a period of 4 weeks and 10 mg/kg b.wt. for another 1 week (total: 5 weeks). 23 Previously, NDEA was given as a single large dose; however, in the current study, smaller doses were given daily throughout the experimental period to reduce the mortality rate. 20 mg/kg was administered at the beginning to initiate hepatocarcinogenesis, followed by 10 mg/kg for the promotion of liver cancer.24 3) Doxorubicin-treated group (HCC+Doxo) in which the rats were intraperitoneally (i.p.) treated with doxorubicin as reference drug in dose of 0.72 mg/rat which is equivalent to the human dose of 20 mg/m2 according to Barnes and Paget25 once weakly for 9 weeks. 4) Punica granatum (high dose)-treated group (HCC+Punica granatum H) in which the rats were orally administered with 2.25 g/kg.b.wt/day of Punica granatum peel extract five times a week during a period of 9 weeks and 5) Punica granatum (low dose)-treated group (HCC+Punica granatum L) in which the rats were orally administered with 1.125 g/kg.b.wt/day of Punica granatum peel extract five times a week during a period of 9 weeks. Blood Sampling After the completion of this round (14 weeks), all animals were fasted for 12 hours then the blood samples were collected from the retro-orbital venous plexus under diethyl ether anaesthesia. Blood samples were left to clot and the sera were separated by cooling centrifugation (4°C) at 1800 xg for 10 min. and stored immediately at 20°C in clean plastic Eppendorf pendng biochemical analysis. Liver Tissue Sampling After blood collection, the animals were sacrificed by cervical decapitation, dissected and the whole liver of each rat was rapidly excised and thoroughly washed with isotonic saline. The whole liver of each rat was divided into three portions; the first and second portions were fixed in formal saline (10%) for 24 h for International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 122 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 immunohistochemical investigation and histological examination respectively whereas, the third portion was snap-frozen directly in liquid nitrogen and stored at -80°C prior to RNA isolation for gene expression analysis. Methods Biochemical Analyses Serum aspartate transaminase (AST) andalanine transaminase (ALT) activity was estimated using Quimica Clinica Aplicada S.A (Spain) kit according to the method 26 described by Reitman and Frankel. Serumalkaline phosphatase (ALP) activity was measured using Quimica Clinica Aplicada S.A.(Spain) kit according to the method 27 described by Bowers and Mc Comb. Serum alphafetoprotein (AFP) was determined by ELISA using kit purchased from Glory Science Co., Ltd (USA), according to the manufacturer's instructions provided with AFP assay kit. Serum carcinoembryonic antigen (CEA) was quantified by ELISA using kit purchased from Glory Science Co., Ltd (USA), according to the manufacturer’s instructions provided with CEA assay kit. Serum glypican-3 (GPC-3) was detected by ELISA using kit purchased from Glory Science Co., Ltd (USA), according to the manufacturer’s instructions provided with GPC-3 assay kit. Serum vascular endothelial growth factor (VEGF) was determined by ELISA using kit purchased from Glory Science Co., Ltd (USA), according to the manufacturer’s instructions provided with VEGF assay kit. Serum hepatocyte growth factor (HGF) was assayed by ELISA using kit purchased from Glory Science Co., Ltd (USA), according to the manufacturer’s instructions provided with HGF assay kit. Serum epidermal growth factor (EGF) was measured by ELISA using kit purchased from Glory Science Co., Ltd (USA), according to the manufacturer’s instructions provided with EGF assay kit. Molecular Genetic Analysis Isolation of Total RNA Total RNA was isolated from liver tissue of female rats by the standard TRIzol® Reagent extraction method (Invitrogen, USA). Briefly, tissue samples were homogenized in 1 ml of TRIzol® Reagent per 50 mg of the tissue. After wards, the homogenized sample was incubated for 15 minutes at room temperature. A volume of 0.2 ml of chloroform per 1 ml of TRIzol® Reagent was added. Then the samples were shaked vigorously by vortex for 15 seconds and incubated at room temperature for 3 minutes. The samples were centrifuged at 12,000 x g for 15 minutes at 4 C. Following centrifugation, the mixture was separated into lower red, phenol-chloroform phase, an interphase, and a colorless upper aqueous phase. RNA was remained exclusively in the aqueous phase. Therefore, the upper aqueous phase was carefully transferred without disturbing the interphase into a clean tube. The RNA was precipitated from the aqueous phase by mixing with isopropyl alcohol. A volume of 0.5 ml of isopropyl alcohol was added per 1 ml of TRIzol® Reagent was used for the initial ISSN 0976 – 044X homogenization. Afterwards, the samples were incubated at 15 to 30 C for 10 minutes and centrifuged at 12,000 x g for 10 minutes at 4 C. The RNA was precipitated forming a gel-like pellet on the side and bottom of the tube. The supernatant was removed completely and the RNA pellet was washed once with 1 ml of 75% ethanol. The samples were mixed by vortex and centrifuged at 7,500 x g for 5 minutes at 4 C. The supernatant was removed and RNA pellet was airdried for 10 minutes. RNA was dissolved in diethylpyrocarbonate (DEPC)-treated water by passing solution a few times through a pipette tip. Total RNA was treated with 1 U of RQ1 RNAse-free DNAse (Invitrogen, USA) to digest DNA residues and resuspended in DEPC-treated water. Purity of total RNA was assessed by 260/280 nm ratio (between 1.8 and 2.1). Additionally, integrity was assured with ethidium bromide-stain analysis of 28S and 18S bands by formaldehyde-containing agarose gel electrophoresis. Aliquots were used immediately for reverse transcription (RT). Reverse Transcription (RT) Reaction The complete Poly(A)+ RNA isolated from female rat liver tissues was reverse transcribed into cDNA in a total volume of 20 µl using RevertAidTM First Strand cDNA Synthesis Kit (MBI Fermentas, Germany). An amount of total RNA (5µg) was used with a reaction mixture, termed as master mix (MM). The MM consisted of 50 mM MgCl2,5x reverse transcription (RT) buffer (50 mM KCl; 10 mM Tris-HCl; pH 8.3; 10 mM of each dNTP, 50 µM oligodT primer, 20 U ribonuclease inhibitor (50 kDa recombinant enzyme to inhibit RNase activity) and 50U M-MuLV reverse transcriptase. The mixture of each sample was centrifuged for 30 sec at 1000xg and transferred to the thermocycler. The RT reaction was carried out at 25 °C for 10 min, followed by 1 h at 42 °C, and the reaction was stopped by heating for 5 min at 99 °C. Afterwards the reaction tubes containing RT preparations were flash-cooled in an ice chamber until being used for DNA amplification through semiquantitative real time-polymerase chain reaction (sqRTPCR). Semi-Quantitative Real Time-Polymerase Chain Reaction (sqRT-PCR) A Rotor-Gene Q - Qiagen (USA) was used to determine the rats cDNA copy number. PCR reactions were set up in 25 L reaction mixtures containing 12.5 L 1× SYBR® Premix Ex TaqTM (TaKaRa, Biotech. Co. Ltd.), 0.5 L 0.2 M forward primer, 0.5 L 0.2 M reverse primer, 6.5 L distilled water, and 5 L of cDNA template. The reaction conditions were summarized in Table 1. Each experiment included a distilled water control. The semi quantitative values of RT-PCR (sqRT-PCR) of the studied genes were normalized on the expression value of ß-actin gene. International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 123 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 Primer sequences were as follows: ING3, F: 5′- agt ggc agg aag agc aaa aa-3′, R: 5′- tcc cac cat ctc tcc ata gg-3′ (GenBank NCBI Reference: NM_001034107.1), Foxp-1, F: 5'- CAG GCA GAT CCC CTA TGC AA-3', R: 5'-GGA CAG AGG 28 GCC TTC AGC TT-3’ and ß-actin, F: 5′- CCC CAT CGA GCA CGG TAT TG-3′, R: 5′-ATG GCG GGG GTG TTG AAG GTC3’.29 At the end of each sqRT-PCR a melting curve analysis was performed at 95.0 °C to check the quality of the used primers. Table 1: sqRT-PCR Reaction Program Temperature Time Step 1: (1 Cycle) a: Intial denaturation step 95.0 °C 03.00 min. Step 2: (30 cycles) a: denaturation step 95.0°C 15 sec. b: annealing step c: extension step 55.0 °C 72.0 °C 00.30 sec. 00.30 sec. Step 3: (71 cycles) a: melting curve analysis 60.0 °C 00.10 sec. Then increased about 0.5 °C every 10 sec. up to 95 °C Calculation of Gene Expression The amplification efficiency (Ef) was calculated from the slope of the standard curve using the following formula:30 Ef = 10-1/slope Efficiency (%) = (Ef – 1) x 100 The relative quantification of the target gene to the reference gene was determined by using the ΔCT method if E for the target (ING3 and Foxp-1) and the reference primers (β-actin) are the same: Ratio (reference/ target gene) = EfCT (reference) – CT (target) Immunohistochemical Investigation After fixation of liver specimens (first portion) in 10% formalin saline for 24 hours, washing was done in tap water then, ascending grade of ethyl alcohol (30%, 50%, 70%, 90% and absolute) were used for dehydration. Specimens were cleared in xylene and embedded in paraffin (melting point 58-60oC) for 24 hours. Sections were cut into 4µ thick by slidge microtome then, fixed on positive slides in a 65 oC oven for 1 hr. Slides were placed in a coplin jar filled with 200 mL of triology working solution (Cell Marque, CA-USA, cat.#920P-04) which combines the three pretreatment steps: deparaffinization, rehydration and antigen unmasking. Then, the jar is securely positioned in the autoclave which was adjusted so that temperature reached 120 oC and maintained stable for 15 min after which pressure is released. Thereafter, the coplin jar is removed to allow slides to cool for 30 min. Sections were then washed and immersed in Tris-buffer saline (TBS) to adjust the pH and these were repeated between each step of the immunohistochemical procedure. Quenching endogenous peroxidase activity was performed by immersing slides in ISSN 0976 – 044X 3% hydrogen peroxide for 10 min. Broad spectrum LABSA detection system (Invitrogen, USA, cat.#85-8943) was used to visualize any antigen-antibody reaction in the tissue. Background staining was blocked by putting 3 drops of 10% goat non immuno serum blocker on each slide and incubating the slides in a humidity chamber for 10 min. Without washing, excess serum was drained and the working solution of the primary antibodies of β-catenin, survivin and Ki-67 mouse monoclonal (Thermo Scientific, USA, cat.#RM-2101-R7 for β-catenin, cat.#RB-9245-P1 for survvin and cat.#RB-9043-R1 for Ki-67) were prepared. Three drops of the working solution were applied. Then, the slides were incubated in the humidity chamber overnight at 4 oC. Hence forward, biotinylated secondary antibody from ultravision detection system antipolyvalent HRP/DAB (Thermo Scientific, USA, cat.#TP015-HD) was applied on each slide for 20 min followed by 20 min incubation with the streptavidin horse radish peroxidase (HRP) enzyme conjugate (Thermo Scientific, USA, cat.#TP-015-HD). Then, 3,3´-diaminobenzidine (DAB) chromogen (Thermo Scientific, USA, cat.#TP-015-HD) was prepared and 3 drops were applied on each slide for 2 min. DAB was rinsed, after which counterstaining with Mayer hematoxylin and cover slipping were performed as the final steps before slides were examined under the light microscope (Olympus Cx21 with attached digital camera).31 Image analysis was performed using the image J, 1.41a NIH, (USA) analyzer. β-catenin and survivin expressions were represented as area percentage of immunopositive cells, while, Ki-67 expression was represented as a count of immunopositive cells. Histological Examination After fixation of the second portion of liver tissue in formal saline (10%) for 24 hours, the tissue was then washed in running tap water, dehydrated in ascending grade of ethyl (30%, 50%, 70%, 90% and absolute alcohol). After that, specimens were cleared in xylene and embedded in paraffin at 56 degree in hot air oven for twenty four hours. Paraffin bees wax tissue blocks were prepared for sectioning at 5µm thick by slidge microtome. The obtained tissue sections were mounted on clean glass slides. The slides were deparaffinized in xylol and then immersed in descending series of alcohol then, ordinary hematoxylin and eosin stain was used to stain the slides before examination under the light microscope (Olympus Cx21 with attached digital camera).32 Statistical Analysis In the present study, the results were expressed as Mean + S.E of the mean. Data were analyzed by one way analysis of variance (ANOVA) using the Statistical Package for the Social Sciences (SPSS) program, version 14 followed by least significant difference (LSD) to compare 33 significance between groups. Difference was considered significant when P value was < 0.05. International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 124 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 RESULTS Biochemical Analyses The data depicted in Table 2 illustrated the effect of Punica granatum peel extract and doxorubicin treatment on the activity of liver enzymes (AST, ALT and ALP) in serum of rats bearing hepatocellular carcinoma (HCC). The present results demonstrated that there is significant increase (P<0.05) in serum AST, ALT and ALP activity in HCC group as compared to the negative control group. Insignificant change (P>0.05) in serum AST, ALT and ALP activity has been detected upon treatment of HCC with doxorubicin when compared with the untreated HCC bearing rats (HCC group). Meanwhile, the treatment of rats bearing HCC with either high (2.25g/kg.b.wt.) or low (1.125g/kg.b.wt.) dose of Punica granatum peel extract elicited significant reduction (P <0.05) in serum AST, ALT and ALP enzymes activity relative to the untreated HCC bearing rats (HCC group). In comparison with doxorubicin-treated group, Punica granatum peel extract either high (2.25g/kg.b.wt.) or low (1.125g/kg.b.wt.) dose produced significant decrease (P< 0.05) in liver enzymes (AST, ALT and ALP) activity in serum of the treated rats (Table 2). Table 2: Effect of treatment with Punica granatum peel extract and doxorubicin on serum AST, ALT and ALP activity in hepatocellular carcinoma rat model Parameters Groups AST (U/L) ALT (U/L) ALP (U/L) Negative control 42.8 ± 0.6 26.7 ± 0.5 145.7 ± 4.3 HCC 80.3 ± 1.5 a 50.1 ± 2.4 a 284.1 ± 6.6 HCC + Doxo 78.6 ± 2.2 47.5 ± 0.7 HCC + Punica granatum H 65.2 ± 1.3 HCC +Punica granatum L 60.1 ± 1.4 bc bc 42.8 ± 0.4 38.3 ± 0.5 bc bc a 279.5 ± 13.3 235.3 ± 4.1 215.3 ± 2.4 bc bc HCC: hepatocellular carcinoma. ISSN 0976 – 044X detected in HCC bearing rats treated with doxorubicin relative to the untreated HCC bearing rats (Table 3). Similarly, the treatment of HCC bearing rats with high dose of Punica granatum peel extract (2.25g/kg.b.wt.) produced insignificant decrease (P > 0.05) in serum AFP level as compared to the untreated HCC bearing rats. Whereas, significant reduction (P < 0.05) in serum CEA and GPC-3 levels has been recorded in HCC bearing rats treated with high dose of Punica granatum peel extract relative to the untreated HCC bearing rats (Table 3). Treatment of rats bearing HCC with low dose Punica granatum peel extract (1.125g/kg.b.wt) showed significant reduction (P< 0.05) in serum AFP, CEA and GPC-3 levels vesus the untreated HCC bearing rats. Also, the data in Table 3 revealed that the treatment of rats bearing HCC with high dose of Punica granatum peel extract elicited insignificant reduction (P> 0.05) in serum AFP, CEA and GPC-3 levels, in comparison with doxorubicin-treated group. Treatment of HCC bearing rats with low dose of Punica granatum peel extract induced significant reduction (P<0.05) in serum AFP and GPC-3 levels versus doxorubicin-treated group. Meanwhile, treatment of rats bearing HCC with Punica granatum peel extract (low dose) produced insignificant reduction (P> 0.05) in serum CEA level in comparison with doxorubicintreated group (Table 3). Table 3: Effect of treatment with Punica granatum peel extract and doxorubicin on serum AFP, CEA and GPC-3 levels in hepatocellular carcinoma rat model Parameters Groups AFP (ng/mL) CEA (ng/mL) Negative control 19.1 ± 1.5 0.12 ± 0.01 9.4 ± 0.2 a a 28.6 ± 0.6 0.6 ± 0.06 GPC-3 (pg/mL) 40.1 ± 2.9 HCC + Doxo 36.9 ± 1.8 0.45 ± 0.04 b 22.4 ± 0.3 b HCC + Punica granatum H 35.9 ± 2.4 0.44 ± 0.02 b 21.8 ± 0.4 b 0.40 ± 0.03 b 18.9 ± 0.3 Doxo: Doxorubicin. HCC + Punica granatum L Punica granatum H: Punica granatum peel extract (high dose). HCC: hepatocellular carcinoma. Punica granatum L: Punica granatum peel extract (low dose). Doxo: Doxorubicin. a: Significant change at P < 0.05 in comparison with the negative control group. Punica granatum H: Punica granatum peel extract (high dose). b: Significant change at P < 0.05 in comparison with HCC group. c: Significant change at P < 0.05 in comparison with HCC + Doxo group. The results in Table 3 represented the effect of Punica granatum peel extract and doxorubicin treatment on serum levels of AFP, CEA and GPC-3 in rats bearing hepatocellular carcinoma (HCC). The current results revealed that there is significant increase (P<0.05) in serum AFP, CEA and GPC-3 levels in HCC group versus the negative control group. Insignificant decrease (P>0.05) in serum AFP level has been recorded in HCC bearing rats treated with doxorubicin as compared to untreated HCC bearing rats. Significant decrease (P<0.05) in serum CEA and GPC-3 levels has been a HCC 30.3 ± 2.2 bc bc Punica granatum L: Punica granatum peel extract (low dose). a: Significant change at P < 0.05 in comparison with the negative control group. b: Significant change at P < 0.05 in comparison with HCC group. c: Significant change at P < 0.05 in comparison with HCC + Doxo group. The results presented in Table 4 revealed the effect of Punica granatum peel extract and doxorubicin treatment on serum levels of VEGF, HGF and EGF in rats bearing hepatocellular carcinoma (HCC). The present results demonstrated that there is significant increase (P<0.05) in serum levels of VEGF, HGF and EGF in HCC group as compared to the negative control group. Treatment of rats bearing HCC with doxorubicin resulted in significant decline (P < 0.05) in serum VEGF and HGF levels as International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 125 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 compared to the untreated HCC bearing rats. Insignificant depletion (P>0.05) in serum EGF level has been recorded in HCC bearing rats treated with doxorubicin with respect to the untreated HCC bearing rats (Table 4). Also, the data in Table 4 revealed that the treatment of HCC bearing rats with Punica granatum peel extract either high dose (2.25g/kg. b. wt.) or low dose (1.125g/kg. b. wt.) elicited significant reduction (P< 0.05) in serum VEGF and HGF levels relative to the untreated HCC bearing rats. Meanwhile, treatment of rats bearing HCC with high dose of Punica granatum peel extract produced insignificant suppression (P> 0.05) in serum EGF level as compared to the untreated HCC bearing rats. However, the treatment of HCC bearing rats with low dose of Punica granatum peel extract led to significant depletion (P< 0.05) in serum EGF level with respect to the untreated HCC bearing rats (Table 4). In comparison with doxorubicin-treated group, the present data showed that the treatment of rats bearing HCC with either high dose or low dose of Punica granatum peel extract produces insignificant depression (P> 0.05) in serum VEGF and EGF levels. Meanwhile, the treatment of rats bearing HCC with either high dose or low dose of Punica granatum peel extract resulted in significant reduction (P< 0.05) in serum HGF level relative to the doxorubicin-treated group (Table 4). Table 4: Effect of treatment with Punica granatum peel extract and doxorubicin on serum VEGF, HGF and EGF levels in hepatocellular carcinoma rat model Parameters VEGF HGF EGF Groups (pg/mL) (ng/mL) (ng/L) Negative control 6.0 ± 0.2 4.2 ± 0.05 a HCC 18.0 ± 0.97 HCC + Doxo 14.4 ± 0.5 b HCC + Punica granatum H 14.1 ± 1.0 b HCC + Punica granatum L 13.1 ± 0.98 b 28.8 ± 0.5 2425 ± 123 16.9 ± 0.3 b 2197 ± 87.7 16.0 ± 0.3 bc 2168 ± 70.5 14.8 ± 0.2 bc 2111 ± 102.5 bearing HCC and treated with doxorubicin versus the untreated HCC bearing rats (Figure 1). Similarly, the treatment of rats bearing HCC with Punica granatum peel extract elicited significant upergulation (P< 0.05) in expression level of ING3 gene in the liver tissue as compared to the untreated HCC bearing rats. In comparison with doxorubicin-treated group, treatment of rats bearing HCC with high dose of Punica granatum peel extract resulted in insignificant change (P> 0.05) in the expression level of ING3 gene in the liver tissue, whereas, significant upregulation (P< 0.05) in the expression level of ING3gene in the liver tissue has been detected in HCC bearing rats treated with low dose of Punica granatum peel extract as compared to doxorubicin-treated group (Figure 1). Figure 1: Effect of treatment with Punica granatum peel extract and doxorubicin on hepatic ING3 gene expression level in hepatocellular carcinoma rat model. HCC: hepatocellular carcinoma. Doxo: Doxorubicin. Punica granatum H: Punica granatum peel extract (high dose). Punica granatum L: Punica granatum peel extract (low dose). a: Significant change at P < 0.05 in comparison with the negative control group. b: Significant change at P < 0.05 in comparison with HCC group. c: Significant change at P < 0.05 in comparison with HCC + Doxo group. 1900 ± 53.5 a ISSN 0976 – 044X a b HCC: hepatocellular carcinoma. Doxo: Doxorubicin. Punica granatum H: Punica granatum peel extract (high dose). Punica granatum L: Punica granatum peel extract (low dose). a: Significant change at P < 0.05 in comparison with the negative control group. b: Significant change at P < 0.05 in comparison with HCC group. c: Significant change at P < 0.05 in comparison with HCC + Doxo group. Molecular Genetic Analysis Figure 1 illustrated the effect of treatment with Punica granatum peel extract and doxorubicin on the expression level of ING3 gene in the liver tissue of rats bearing hepatocellular carcinoma. The current results revealed that there is significant down regulation (P<0.05) in the expression level of ING3 gene in the liver tissue of rats bearing HCC when compared with the negative control group. Significant up regulation (P<0.05) in the expression level of ING3 gene has been detected in liver tissue of rats Figure 2: Effect of treatment with Punica granatum peel extract and doxorubicin on hepatic Foxp-1 gene expression level in hepatocellular carcinoma rat model. HCC: hepatocellular carcinoma. Doxo: Doxorubicin. Punica granatum H: Punica granatum peel extract (high dose). Punica granatum L: Punica granatum peel extract (low dose). a: Significant change at P < 0.05 in comparison with the negative control group. b: Significant change at P < 0.05 in comparison with HCC group. c: Significant change at P < 0.05 in comparison with HCC + Doxo group. International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 126 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 Figure 2 represented the effect of treatment with Punica granatum peel extract and doxorubicin on the expression of Foxp-1 gene in the liver tissue of rats bearing hepatocellular carcinoma. The present results showed that there is significant upregulation (P<0.05) in the expression level of Foxp-1 gene in the liver tissue of rats bearing HCC with respect to the negative control group. Treatment of HCC bearing rats with doxorubicin elicited significant downregulation (P<0.05) in the gene expression level of Foxp-1 in the liver tissue versus the untreated of HCC bearing rats (Figure 2). In the same manner, the treatment of rats bearing HCC with either high dose or low dose of Punica granatum peel extract induced significant downregulation (P< 0.05) in the expression level of Foxp-1 gene in the liver tissue in comparison with the untreated HCC bearing rats. In comparison with doxorubicin-treated group, the treatment of rats bearing HCC with high dose of Punica granatum peel extract produced insignificant downregulation (P> 0.05) in the expression level of Foxp1 gene in the liver tissue. Whereas, significant downregulation (P< 0.05) in the expression level of Foxp-1 gene in the liver tissue was recorded in HCC bearing rats treated with low dose of Punica granatum peel extract as compared to doxorubicin-treated group (Figure 2). ISSN 0976 – 044X Immunohistochemical Investigation In the present study, the expression of β-catenin, survivin and Ki-67 in liver tissue of the different experimental groups has been investigated by immunohistochemical technique. Immunohistochemical staining of liver tissue section of rat in the negative control group using antibody against β-catenin showed area percentage of β-catenin immunopositive cells (3.50 %) (Figure 3). While, immunohistochemical staining of liver tissue section of rat in HCC group using antibody against βcatenin showed sever increase in the area percentage of β-catenin immunopositive cells (30.9 %) (Figure 4). Immunohistochemical staining of liver tissue section of rat bearing HCC and treated with doxorubicin using antibody against β-catenin showed mild decrease in the area percentage of β-catenin immunopositive cells (23.3 %) (Figure 5). Immunohistochemical staining of liver tissue section of rat bearing HCC and treated with high dose of Punica granatum peel extract using antibody against β-catenin showed moderate decrease in the area percentage of βcatenin immunopositive cells (16.4 %) (Figure 6). Immunohistochemical staining of liver tissue section of rat bearing HCC and treated with low dose of Punica granatum peel extract using antibody against β-catenin showed marked decrease in the area percentage of βcatenin immunopositive cells (7.4 %) (Figure 7). Figure 3: Photomicrograph for staining of liver tissue section of rat in the negative control group using antibody against β-catenin (80x). Figure 4: Photomicrograph for staining of liver tissue section of rat in HCC group using antibody against βcatenin (80x). Figure 5: Photomicrograph for staining of liver tissue section of rat bearing HCC and treated with doxorubicin using antibody against β-catenin (80x). Figure 6: Photomicrograph for staining of liver tissue section of rat bearing HCC and treated with high dose of Punica granatum peel extract using antibody against βcatenin (80x). International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 127 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 ISSN 0976 – 044X Figure 7: Photomicrograph for staining of liver tissue section of rat bearing HCC and treated with low dose of Punica granatum peel extract using antibody against βcatenin (80x). Figure 8: Photomicrograph for staining of liver tissue section of rat in the negative control group using antibody against survivin (80x). Figure 9: Photomicrograph for staining of liver tissue section of rat in HCC group using antibody against survivin (80x). Figure 10: Photomicrograph for staining of liver tissue section of rat bearing HCC and treated with doxorubicin using antibody against survivin (80x). Figure 11: Photomicrograph for staining of liver tissue section of rat bearing HCC and treated with high dose of Punica granatum peel extract using antibody against survivin (80x). Figure 12: Photomicrograph for staining of liver tissue section of rat bearing HCC and treated with low dose of Punica granatum peel extract using antibody against survivin (80x). Figure 13: Photomicrograph for staining of liver tissue section of rat in the negative control group using antibody against Ki-67 (80x). Figure 14: Photomicrograph for staining of liver tissue section of rat in HCC group using antibody against Ki-67 (80x). International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 128 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 Figure 15: Photomicrograph for staining of liver tissue section of rat bearing HCC and treated with doxorubicin using antibody against Ki-67 (80x). ISSN 0976 – 044X Figure 16: Photomicrograph for staining of liver tissue section of rat bearing HCC and treated with high dose of Punica granatum peel extract using antibody against Ki-67 (80x). Figure 17: Photomicrograph for staining of liver tissue section of rat bearing HCC and treated with low dose of Punica granatum peel extract using antibody against Ki-67 (80x). C B A E D Figure 18: Photomicrograph of liver tissue section of rat in (A): The negative control group showing normal histological structure of the central vein (cv) and the surrounding hepatocytes (h) (H&E x 40), (B): HCC group showing anaplastic hepatocytes activities (h) charcacterized by polarity, pleomorphism and hyperchromachtic nuclei and the ratio between the nucleus and cytoplasm was 1:1 associated with acinar glandular structure formation (a) (H&E x 40), (C): HCC group and treated with doxorubicin showing thickening and fibrosis (f) with inflammatory cell infiltration (m) and dilated blood capillanes (v) in hepatic capsule. Also, many hepatocytes showed karomegaly and pyknotic nuclei (H&E x 40), (D): HCC group and treated with high dose of Punica granatum peel extract showing inflammatory cells infiltration in the portal area (m) with sever dilatation of in the portal vein (cv) and focal degenerated hepatocytes (d) (H&E x 40) and (E): HCC group and treated with low dose of Punica granatum peel extract showing proliferative cystic bile ducts (bd) (H&E x 16). Immunohistochemical staining of liver tissue section of rat in the negative control group using antibody against survivin showed area percentage of survivin immunopositive cells (1.49 %) (Figure 8). Immunohistochemical staining of liver tissue section of rat in HCC group using antibody against survivin showed sever increase in the area percentage of survivin immunopositive cells (26.18 %) (Figure 9). Immunohistochemical staining of liver tissue section of rat bearing HCC and treated with doxorubicin using International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 129 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 antibody against survivin showed mild decrease in the area percentage of survivin immunopositive cells (20.51 %) (Figure 10). Immunohistochemical staining of liver tissue section of rat bearing HCC and treated with high dose of Punica granatum peel extract using antibody against survivin showed moderate decrease in the area percentage of survivin immunopositive cells (14.39 %) (Figure 11). Immunohistochemical staining of liver tissue section of rat bearing HCC and treated with low dose of Punica granatum peel extract using antibody against survivin showed moderate decrease in the area percentage of survivin immunopositive cells (6.76 %) (Figure 12). Immunohistochemical staining of liver tissue section of rat in the negative control group using antibody against Ki-67 showed negative reaction (the number of immunopositive cells for Ki-67 expression was zero cell) (Figure 13). Immunohistochemical staining of liver tissue section of rat in HCC group using antibody against Ki-67 showed sever increase in the number of immunopositive cells for Ki-67 expression (65 cells) (Figure 14). Immunohistochemical staining of liver tissue section of rat bearing HCC and treated with doxorubicin using antibody against Ki-67 showed mild decrease in the number of immunopositive cells for Ki-67 expression (37 cells) (Figure 15). Immunohistochemical staining of liver tissue section of rat bearing HCC and treated with high dose of Punica granatum peel extract using antibody against Ki-67 showed mild decrease in the number of immunopositive cells for Ki-67 expression (28 cells) (Figure 16). Immunohistochemical staining of liver tissue section of rat bearing HCC and treated with low dose of Punica granatum peel extract using antibody against Ki67 showed marked decrease in the number of immunopositive cells for Ki-67 expression (5 cells) (Figure 17). Histological Examination Histological examination of liver tissue section of rat in the negative control group stained with hematoxylin and eosin (H&E) showed normal histological structure of the central vein and the surrounding hepatocytes and no histopathological alteration was observed (Figure 18 A). While, microscopic investigation of liver tissue section of rat in HCC group showed anaplastic hepatocytes activities indicated by polarity, pleomorphism and hyperchromachtic nuclei and the ratio between the nucleus and cytoplasm was 1:1 associated with acinar glandular structure formation (Figure 18 B). Photomicrograph of liver tissue section of rat bearing HCC and treated with doxorubicin showed thickening and fibrosis with inflammatory cell infiltration and dilated blood capillanes in hepatic capsule. Also, many hepatocytes showed karomegaly and pyknotic nuclei (Figure 18 C). Photomicrograph of liver tissue section of rat bearing HCC and treated with high dose of Punica granatum peel extract showed inflammatory cells ISSN 0976 – 044X infiltration in the portal area with sever dilatation of in the portal vein and focal degenerated hepatocytes (Figure 18 D). Photomicrograph of liver tissue section of rat bearing HCC and treated with low dose of Punica granatum peel extract showed proliferative cystic bile ducts (Figure 18 E). DISCUSSION This study was constructed to elucidate the underlying biochemical and molecular mechanisms by which Nnitrosodiethylamine could induce hepatocellular carcinoma in rats. Also, the aim of the present study was extended to explore the possible therapeutic potential of Punica granatum peel extract against hepatocellular carcinoma with special concern on its mode of action. These goals were achieved through conducting liver function tests, relevant tumor and angiogenic as well as genetic markers, differentiated, antiapoptotic and proliferative indices. A model close to human situation is the chemical induction of HCC with special carcinogens such as Nnitrosodiethylamine (NDEA). As it is widely accepted that metabolic activation of nitrosamines by cytochrome P450 enzymes to reactive electrophiles are required for their cytotoxic, mutagenic and carcinogenic activity. Because of its relatively simple metabolic pathway and potent carcinogenicity, NDEA has been used as an effective experimental model in the field of carcinogenesis and chemoprevention.34 The data of the present study showed that NDEA administration led to significant increase in liver enzymes activity AST, ALT and ALP in serum. These findings are in agreement with Willimsky35 Pratt and Kaplan36 reported that these enzymes are commonly elevated in patients with liver diseases and the elevated values of these enzymes in serum reflect the status of liver injury. NDEA is mainly metabolized in the liver by the action of cytochrome p450 enzymes and its reactive metabolites are primarily responsible for its hepatotoxic effects. NDEA is bioactivated to ethyldiazonium ion which alkylates DNA bases to form pro-mutagenic adducts such as O6ethyldeoxyguanosine and O4 and O6ethyldeoxythymidine.These bioactive molecules has the ability to induce oxidative stress and hepatotoxicity by damaging the biomolecules such as DNA, lipids and proteins.12 The observed increment of liver enzymes in serum as a consequence of NDEA administration could be due to the production of free radicals during NDEA metabolism, which damage the hepatocellular membrane. As a result, these cytoplasmic enzymes are 37 released into the systemic circulation. The current results revealed that the treatment of rats bearing HCC with Punica granatum peel extract produced significant decrease in serum AST, ALT and ALP enzymes activity. These findings are greatly supported by those of 38 Rao and Dama who reported that Punica granatum decreased the enzymatic activity of liver enzymes in International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 130 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 serum and alleviated the degree of liver damage. These effects might be related to that Punica granatum extract exhibits strong antioxidant activity, scavenges free radicals and ameliorates the damaging impact on the 39 liver, due to its high polyphenolic contents including ellagic acid and ellagitannins.40 The inhibitory effect of Punica granatum on lipid peroxidation in rat liver microsomes is related to the ability of its phenolic and flavonoid compounds mainly ellagic acid41,42 to inhibit 16,39 hepatic oxidative enzymes (cytochrome P450 system). Moreover, Abhijeet and Rupali43 stated that Punica granatum possessed potential hepatoprotective activity which might be attributed to its flavonoids (anthocyanidins) content. Punica granatum peel extract has been found to have a potent antioxidant activity due to its active compounds that are electron donors and can react with free radicals to convert them to more stable products and thus terminate radical chain reaction.39,44 This indicates that Punica granatum peel extract could preserve the structural integrity of the hepatocellular membrane and liver cell architecture. Therefore, it could prevent the leakage of liver enzymes into serum and restore liver functions.45 The present data revealed that serum AFP level in rats bearing HCC displayed significant elevation. This finding comes in line with that of Sadiknah46 and Sivaramakrishnan47 who reported that rats injected with NDEA experienced significant increase in serum AFP level. It has been found a high AFP gene expression level in liver tissue of rats administered with NDEA.48 Moreover, Zhou49 demonstrated that the high serum concentration of AFP in HCC patients might be due to the tumor excretion of this protein. Furthermore, some clinical researches have indicated that the high percentage of AFP is closely related to poor differentiation and biologically malignant characteristics especially portal vein invasion of HCC.50,51 The possible explanations for the reinitiation of AFP synthesis by neoplastic hepatocytes include either increased transcription of AFP gene or post-translational modification affecting AFP production. In rats which have been exposed to chemical carcinogens or those with HCC, AFP production is roughly proportional to the amount of transplantable mRNA present.52 Steady state AFP transcription may begin prior to the development of histologically or symptomatic hepatocellular carcinoma.53 In view of the present data serum CEA level in rats bearing HCC showed significant increase. The increase in serum CEA levels upon NDEA administration was presumably associated with production rates of tumor, its 54 location and stage, size, differentiation and vascularity. 55 Macnab demonstrated that the tumor could cause a release of CEA from damaged liver cells adjacent to it. Experimental studies have shown that CEA is normally 56 removed from circulating plasma by the liver. Thus, the increased serum levels of CEA might therefore results ISSN 0976 – 044X from impaired hepatic uptake of CEA or CEA-like glycoproteins. It is well known that varying degrees of liver dysfunction occur in patients with HCC. The current results revealed that NDEA administration led to significant increase in serum glypican-3 (GPC-3) level. Glypican-3 is a heparan sulfate proteoglycan that has an important role in cell growth and differentiation, and its function in tumorigenesis is tissue-dependent.57 Moreover, the expression of GPC3, its receptor, and other growth factors coordinate signal transduction pathways that regulate cellular morphology and a variety of cellular behaviors, such as adhesion, proliferation, migration, survival, and differentiation. GPC-3 is differentially expressed during the invasive growth of liver cancer, suggesting that its expression might be involved in the initiation of liver cancer, and might be a critical step in liver cancer development.58,59 Capurro60 recorded overexpression of GPC-3 mRNA and protein in both serum and tissues of early-stage HCC. Also, Capurro61 stated that GPC-3 could stimulate the growth of HCC cells by stimulating the Wnt pathway through facilitating the interaction between the Wnts and their signaling receptors. Chan62 suggested that the upregulation of GPC-3 in some HCC tumors might be attributed to c-Myc gene amplification. As, it has been demonstrated that cMyc transcriptionally regulates GPC3 expression by direct interaction with the GPC3 promoter. Together, these data indicate that the transcription factor c-Myc is a novel regulator of GPC3 gene and may accelerate the proliferation of liver cancer cells at least in part by upregulating the GPC3 pathway. Moreover, the exogenous overexpression of c-Myc increased the endogenous mRNA and protein levels of GPC3.57 In accordance with our results, Suzuki63 indicated that circulating levels of GPC-3 protein were significantly elevated in patient with HCC and the expression of mRNAs for GPC-3 was markedly elevated in the liver carcinoma tissues. Therefore, GPC-3 showed potential as a tumor biomarker for HCC that can be used molecularly targeted therapy. Signal transducer and activators of transcription 3 (STAT3) is involved in oncogenesis through the upregulation of genes encoding apoptosis inhibitors [Bcl-xL, Mcl-1(Bcl-xL as well as Mcl-1 are a Bcl-2 family protein which can act as an apical molecule in apoptosis) and survivin], cellcycle regulators (cyclin D1 and c-Myc (DNA binding protein concerning the regulation on metabolism of nucleic acid and the response to growth factor)), and inducers of angiogenesis (vascular endothelial growth factor). It has been found that STAT3 has an indirect role in GPC-3 expression via upregulation of c-myc and the poor control of the pathway mediated by STAT3 plays a 63 role GPC-3 overexpression. Therefore, STAT3 has recently drawn attention as a novel target for cancer 64,65 therapy. In light of the present data, treatment of HCC bearing rats with Punica granatum peel extract significantly decreased International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 131 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 serum AFP, CEA and GPC-3 levels. The observed decline in serum levels of AFP as a consequence of treatment with Punica granatum peel extract might be related to the apoptotic effect exerted by ellagic acid, the active compound in Punica granatum. Ellagic acid can stimulate apoptosis and completely inhibit the proliferating capacity of the tumor by decreasing nuclear factor kappa B (NF-κB) activity, thereby activating the mitochondrial death pathway, which is associated with loss of mitochondrial membrane potential, cytochrome C release, and caspase-3 activation.66 The significant reduction in serum CEA levels in rats bearing HCC and treated with Punica granatum peel extract could be attributed to the modulating effect of ellagic acid on the oxidative stress. Ellagic acid is a naturally existing phenolic antioxidant found in Punica granatum.67 It shows potent antioxidant effects by radical scavenging activity and the inhibition of lipid peroxidation.68,69 Moreover, the anthocyanin, one of the active constituents of Punica granatum extract, is very reactive towards reactive oxygen species (ROS) because 70 of its electron deficiency and thus it can counteract oxidative stress. In addition, the antioxidant activity of Punica granatum might be related to its flavonoids constituents such as kaempferol and naringin.71 The synergism between all of these antioxidant compounds in Punica granatum peel extract could counteract oxidative stress with consequence regression of serum CEA levels. The suppressive effect of Punica granatum peel extract on serum GPC-3 level in HCC bearing rats, could be explained by more than one mechanism. First, the capability of ellagic acid to inhibit cancer cell growth, via reducing the expression of c-Myc72 which is a direct regulator of GPC-3 gene.57 Second, the ability of luteolin to induce cell cycle arrest at G0/G1 phase and apoptosis through increasing Bax/BclXL ratio and activating caspase3 in hepatoma cell lines.73 Luteolin has been found to induce apoptosis in hepatoma HLF cells by inactivating STAT3 protein, a negative regulator of Fas/CD95 signaling, through ubiquitin-dependent degradation and blocking of CDK5-dependent phosphorylation. This leads to the increased function of Fas/CD95 signaling, caspase-8 activation and ultimately apoptosis.74 In fact, the antiapoptotic genes encoding c-Myc, Bcl-2, Bcl-xl, cyclin D1, and survivin are downstream targets of STAT3.75 Therefore, the good control of STAT3 by luteolin present in Punica granatum peel extract resulted in the significant decline in GPC-3 serum level in the treated rats as shown in the present study. The present results revealed that NDEA administration led to significant increase in serum VEGF, HGF and EGF levels. The increase in serum VEGF is greatly supported by 37 76 the finding of El Mesallamy. Moreover, Liu reported that the expression of VEGF in NDEA administered rats was significantly upregulated. This finding might be attributed to the high angiogenic activity of NDEAinduced hepatocellular carcinoma in rats. Hiramoto77 ISSN 0976 – 044X reported that NDEA decomposition is accompanied with concomitant release of NO in contact with reactive oxygen species. Ueno78 suggested that ROS generated by the NDEA could activate the NF-κB-dependent pathways of neutrophil stimulation and enhance proinflammatory cytokine release and NO production. High levels of interleukin-1 (IL-1) or a combination of proinflammatory cytokines, including interleukin-1 (IL-1), tumor necrosis factor (TNF)-α, and interferon (IFN)-γ, could induce iNOS expressionin hepatocytes under a variety of experimental conditions,79,80 the excessive expression of iNOS leads to an accumulation of NO, which participates in cancer 81,82 development. As, the increased NO level in NDEA administered animals has been found to enhance angiogenesis by stimulating the synthesis of VEGF.83,84 VEGF gene promotor contains many regulatory sequences including consensus binding sites for specificity protein1(SP-1), activator protein-1(AP-1) and AP-transcription factors, as well as hypoxia-inducible factor-1(HIF-1).85 The data using tumor cells, have revealed that NO stabilizes H IF-1 subunit leading to higher HIF-1 binding capacity and 86 increased transcriptional activity of VEGF promotor. Moreover, by upregulating hypoxia-inducible factor, hepatocyte growth factor (HGF) expression is increased in tumor and in the surrounding normal interstitial cells in association with increased expression of HGF receptor (cmet) in tumor and the endothelial cells. HGF c-met signaling induces upregulation of tumoral VEGF expression and endothelial VEGFR2 expression and downregulation of the endogenous inhibitors of angiogenesis.87,88 Dual VEGF and c-met axis activity demonstrates the increased capillary formation in vivo, tubulogenesis in vitro and tumoral microvessel density.88 The significant increase in serum HGF level as a consequence of NDEA administration in rats observed in the present study is greatly supported by the finding of Burr.89 HGF is the most potent growth factor for hepatocytes and has been reported to be elevated in a number of liver diseases, including acute and chronic hepatitis, liver cirrhosis, HCC, primary biliary cirrhosis, and 90 hepatic failure, as the expression of HGF is increased in 91 response to liver injury. Circulating pro HGF undergoes proteolytic conversion via extracellular proteases including HGF activator (HGFA).91 Pro-HGFA is activated by thrombin in response to tissue injury and malignant transformation.92,93 Stellate cells and myofibroblasts have been found to be stimulated to secrete HGF by tumor cell products and HGF has been found to stimulate tumor cell invasiveness in turn.94 High c-met (HGF receptor) expression has been shown in invasive-type of HCC and has been associated with metastasis and reduced overall 95,96 survival. Moreover, the elevated HGF levels in HCC patients may reflect the impaired clearance of HGF due to severe liver damage or may be caused by increased HGF 97 production to regenerate hepatocytes. It has been suggested that the elevated serum HGF is probably produced by infiltrating mesenchymal and cancer cells.98 HGF is secreted primarily by mesenchymal International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 132 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 cells or by stellate and endothelial cells in the liver as an inactive precursor (pro HGF).99 HGF transcription is upregulated by inflammatory modulators such as TNF-α, IL-1,transforming growth factor beta (TGF-beta) and VEGF 91,100 in response to liver injury. Based on inflammation and releasing of the proinflammatory cytokines stimulated by NDEA,78 the nuclear transcription factor NFκB controls a number of genes involved in inflammatory responses, cell cycle progression, inhibition of apoptosis and cell adhesion, hence it plays a critical role in promoting carcinogenesis and cancer progression.101 Preiss and Sattar102 reported that NF-κB activation of Kupffer cells, and macrophages within liver tissue increased the expression of HGF and consequently its protein level. NDEA administration in the present work resulted in significant increase in serum EGF level. The principal angiogenic factors include VEGFs, PDGFs, TGF-alpha and beta, basic fibroblast growth factor (FGF), epidermal growth factor (EGF), hepatocyte growth factor (HGF), angiopoietins and interleukin-4 and -8.103,104 It has been shown that some members of the NADPH oxidase (NOX1) family might play essential roles in regulating the autocrine growth and survival of liver tumor cells and this mechanism might be related to over activation of the angiogenic cascade mainly EGFR pathway. Early ROS production by NOX1 activates a Src/ERK pathway that promotes a positive feedback loop on NOX1 upregulation. In parallel, NOX1- induced ROS stimulates p38 and AKT activation which in turn induce TGF-β and EGFR expression with consequent activation of EGFR pathway to induce cell growth. This pathway might be common to tumoral liver cells, being inactive in normal cells.105 Treatment of HCC bearing rats with Punica granatum peel extract elicited significant reduction in serum levels of VEGF, HGF and EGF. The decreased serum level of VEGF as a consequence of Punica granatum treatment might be attributed to its polyphenolic constituent, ellagic acid. Wang106 reported that ellagic acid exerts potent antiangiogenic activity via specifically and directly targeting vascular endothelial growth factor receptor-2 (VEGFR-2) and its signaling pathway. As ellagic acid could inhibit VEGF-induced phosphorylation of VEGFR-2 in endothelial cell as well as platelet-derived growth factorinduced phosphorylation of platelet-derived growth factor receptor (PDGFR) in smooth muscle cells, leading to the inhibition of downstream signaling triggered by these receptors.107 Moreover, another active constituent of Punica granatum called anthocyanidins experienced anticarcinogenic effects through antiangiogenic, 108 antimutagenic activities and inhibition of cyclooxygenase-2 (COX-2) activity and nitric oxide 109 production leading to the reduction of VEGF level. This represents an indirect mechanism for regression of VEGF serum level in HCC bearing rats treated with Punica granatum peel extract. Additionally, recent research efforts have shown the efficacy of kaempferol which is a member of flavonols and abundantly found in Punica ISSN 0976 – 044X 71 granatum extract in impairing cancer angiogenesis both in vitro and in vivo through inhibiting VEGF secretion in human cancer cells. Kaempferol appears to inhibit VEGF expression and angiogenesis through an ERK-NF-κB-cMyc-p21 pathway. Kaempferol administration has been shown to discourage ERK phosphorylation as well as NFκB and c-Myc expression, the reduction of which, promotes p21 expression, which is a tumor suppressor protein known to antagonize VEGF secretion.110 Furthermore, kaempferol exerts its effects on VEGF regulators asit lowers HIF levels, and it blocks AKT phosphorylation, impeding the signaling machinery that 111 calls for increased VEGF production. The observed depletion in serum level of HGF upon treatment of HCC bearing rats with Punica granatum peel extract might be related to the chemopreventive effect exerted by its active phytochemicals. Bhatia112 demonstrated that Punica granatum phytochemicals exerted chemoprevention against hepatocellular carcinoma through antiproliferative and proapoptotic mechanisms via modulating (NF-κB). Luteolin exhibited its antitumor activity by affecting numerous biochemical pathways critical for the regulation of cell survival, apoptosis, angiogenesis and metastasis, including PI3K/Akt, NF-κB, MAPKs, MMPs and E-cadherin.113,114 Luteolin has been shown to inhibit metastasis of HepG2 cells by blocking hepatocyte growth factor (HGF) activity.115 In particular, luteolin has been found to suppress phosphorylation of c-Met, the membrane receptor of HGF, and then inhibits activation of MAPK/ERKs and P13K-Akt pathways involved in HepG2 cell metastasis.115 The recorded decline in serum level of EGF due to treatment of HCC bearing rats with Punica granatum peel extract could be attributed to that both luteolin and quercetin have protein tyrosine kinase (PTK) inhibitory effect and they exert their antitumor effect by mediating epidermal growth factor receptor (EGFR) tyrosine kinase activity as they can inhibit autophosphorylation of 116,117 EGFR. The results of the current study showed that the expression level of inhibitor of growth-3 (ING3) gene in liver tissues of NDEA administered rats was significantly downregulated. The tumor-suppressor ING3 has been shown to be involved in tumor transcriptional regulation, apoptosis and cell cycle arrest. Yang118 demonstrated that the dysregulation of ING3 in HCC occurred at both transcription and translation levels. Downregulation of ING3 was found to be correlated with tumorigenesis and 119 the progression of HCC. It was demonstrated that ING3 was degraded by S-phase kinase-associated protein-2 120 (Skp2) mediated ubiquitin proteasome system. In addition, there was a correlation between the downregulation of ING3 protein and tumor grades as it has been shown that the low expression of ING3 protein is correlated with more aggressive behavior of the tumor.118 The deregulation of ING3 plays a continuous International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 133 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 role throughout the processing of cancer advancement and development. Furthermore, the negative expression of ING3 is also 121 associated with elevated AFP serum levels. It has been reported that p53 negatively regulated AFP gene expression through alteration of chromatin structure at the core promoter.122,123 Studies of Nagashima124 and Luo125 demonstrated that ING3 gene regulated p53. Thus, it is possible to postulate that ING3 could regulate the AFP gene through modifying p53 function. Mutations in p53 gene found in HCC have shown allelic loss at the 17p13.1 region of the chromosome.126,127 ING3 has been shown to modulate p53-mediated transcription, cell cycle control, and apoptosis, possibly by modulating the NuA4 complex histone acetyltransferase activity. Thus, ING3 cooperates with p53 to regulate apoptosis death receptor/extrinsic apoptotic pathways.124 In addition, ING3 has been found to upregulate Fas expression at both mRNA and protein levels.128 In the present study, NDEA administration resulted in overexpression of liver forkhead box transcription factor (Foxp-1) gene level in rats. Foxp-1, a member of F box family of ubiquitously expressed transcription factors, plays a critical role in cancer development.129 Foxp-1 is unique because it can act as an oncogene and tumor suppressor depending upon the tissue type.130 Significant upregulation of Foxp-1 protein in human primary HCC suggests its potential role in hepatic tumorigenesis.131 Foxp-1 has been identified as a potential target of miR-1 and the upregulation of miR-1 target Foxp-1 in hepatocellular carcinomas. This further explains the growth regulatory functions of miR-1 in the liver and probably in other tissues by predisposing these tissues to neoplastic transformation due to loss of miR-1. Recently, several studies have showed the deregulation of miR-1 in many types of tumor, such as hepatocellular,132 prostate,133,134 thyroid,135 bladder136 and renal137 cancer. Numerous observations indicated an epigenetic deregulation of miRNAs expression in different tumors, including HCC. MiR-1 downregulation in human HCC is 131 associated with promoter methylation. Treatment of primary HCC cultures with the hypomethylating agent caused miR-1 reexpression, which in turn led to downregulation of miR-1 oncogenic targets, including FoxP1 which was overexpressed in HCCs, causing inhibition of cell cycle progression and cell survival.131 As miR-1 has a specific targets including Foxp-1 that have roles in tumorigenesis, it is likely that miR-1’s ability to reduce the expression of many tumor promoting genes could have a global influence on the suppression of tumor 131,138-140 development. Treatment of HCC bearing rats with Punica granatum peel extract produced significant upregulation in the expression level of liver ING3 gene. This finding might be attributed to the apoptotic effect exerted by ellagic acid that activates expression of tumor suppressor genes p53 and p21, leading to cell cycle arrest at the G1/S phase and ISSN 0976 – 044X 141,142 apoptosis. Ellagic acid induces G0/G1-phase arrest of the cell cycle by decreasing expression of Cyclindependent kinase 2(CDK2)142 S-phase arrest of the cell cycle by downregulation of the expression of cyclin A and 143 B1 and whole cell cycle arrest by downregulation of Protein kinase C alpha (PKCα)144 and cyclin D1.72 Moreover, Horinaka145 found that luteolin not only induced the mitochondrial pathway of apoptosis but also caspase 8/10 activation and death receptor (DR-5) expression in human malignant cells. These properties of ellagic acid and luteolin in Punica granatum peel extract are of great value in preserving gene expression level of liver ING3. Treatment of HCC bearing rats with Punica granatum peel extract produced significant down regulation in the expression level of liver Foxp-1 gene. Luteolin, the substrate of catechol-Omethyltransferase (COMT),146 has the ability to indirectly inhibit DNA methyltransferase-1 (DNMT-1) by elevating endogenous S-adenosyl homocysteine (SAH) concentration.147 In fact, the indirect inhibitory effect of polyphenols with catechol (luteolin) or galloyl such as epigallocatechin gallate (EGCG) moiety on DNMTs activity has been demonstrated. These compounds could indirectly regulate DNMT activity by regulating the ratio of S-adenosyl methionine (SAM) and (SAH) during their metabolic methylation by COMT.147,148 This means that these compounds are considered as hypomethylating agents that have the ability to cause reexpression of miR-1 with consequent downregulation of its targets genes including Foxp-1 gene. The results of the current study showed that NDEA administration could up-regulate liver expression of βcatenin in rats. The molecular pathogenesis of HCC is multifactorial and is reliant upon dysregulation of multiple pathways including Wnt/β-catenin, mitogen activated protein kinase (MAPK), phosphatidylinositol3kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR), VEGF, PDGF, insulin-like growth factor (IGF), EGF, TGF-β and HGF.149,150 One pathway of critical importance in HCC is the Wnt/β-catenin signaling. β-catenin is the central effector of the canonical Wnt signaling, which is a highly conserved pathway regulating critical cellular processes such as proliferation, differentiation, survival and self-renewal.151,152 In the absence of Wnt, β-catenin is phosphorylated at amino-terminal serine and threonine residues and targeted for ubiquitination.153 Upon binding of Wnt protein to its cell surface receptor Frizzled and coreceptor low density lipoprotein–related protein 5/6 (LRP5/6), a signal is transduced through disheveled that allows the inactivation of degradation complex comprised of glycogen synthase kinase 3b (GSK3b), adenomatous polyposis coli gene product (APC) and casein kinase Ia, which allows β-catenin to dissociate and translocate to the nucleus to bind to lymphoid enhancer-binding factor/T cell factor (LEF/TCF) family of proteins to transactivate target genes. The Wnt/β-catenin pathway has been implicated in a subset of HCC where activating mutations in the β-catenin gene (CTNNB1) have been International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 134 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 154,155 reported in HCC patients. In HCC, accumulation of βcatenin was present in the early stage of HCC.156,157 The previous studies investigated the correlation between βcatenin expression and the differentiation grades of HCC, 158,159 and prognostic roles of β-catenin expression in HCC. 156 Suzuki found β-catenin protein expression was located mainly in the cell membrane of highly differentiated HCC tissue with a low Ki-67 proliferation index. In contrast, cytoplasmic or nuclear β-catenin protein expression was observed in less differentiated HCC tissue with a high proliferation index. Thus it can be hypothesized that, to some extent, the change in localization of β-catenin 156 expression reflects the differentiation of HCC tissues. In view of the present data, NDEA administration led to overexpression of liver survivin in rats. This result comes 160 in line with Zhu Survivin, a novel member of the Inhibitors of apoptosis proteins (IAP) family, inhibits the activation of caspase-3 and -7, which are downstream effectors of apoptosis, in cells exposed to apoptotic stimuli.161 Also, expression of survivin may occur during early malignant transformation or following a disturbance 162 in the balance between cell proliferation and death. Previous studies have shown that survivin is expressed at a high level in 60%-100% of the most common human tumor types, including colon, pancreas, breast, lung, liver, brain, lymphoma, melanoma and prostate cancers.163,164 Many cancer cell lines, including human hepatoma cells, also displayed resistance toward TRAIL-mediated apoptosis.165,166 There is a growing body of evidence indicates that intracellular antiapoptotic pathways including NF-kB and Akt modulate the response to TRAIL in many cancer cells and contribute to resistance toward TRAIL.167,168 NF-kB has been found to be directly activated by TRAIL169-171 and it might upregulate antiapoptotic molecules, including cIAP1, and cIAP-2172 and their members including survivin. In light of the present data, the expression of Ki-67 in liver tissue of rats administered NDEA showed significant upregulation. This result is in agreement with Cui173 who demonstrated that Ki-67 expressing rate in cancer tissue was higher than that in para-cancer tissue. Our results are 174 in corformity with that of Pizem who stated that PCNA and Ki-67 were useful for proliferative activity assessment of hepatocytes and their expressions were higher in HCC than in non-neoplastic liver. These authors demonstrated a statistically significant correlation between PCNA and Ki-67 proliferative indices in HCC as well as a positive correlation between tumor grade and Ki-67 index. Umemura175 reported that high dose of NDEA in experimental animals induced cell proliferation associated with DNA damage, mutations and induction of 176 HCC. On the other hand, study of Yeh on human HCC with histological heterogeneity showed that differentiation of human HCC induced telomerase activation and Ki-67 expression. Tumor cells consist of proliferating cells (S, G2, M, G1 stage), temporarily non-porliferating cells (G0) and non- ISSN 0976 – 044X proliferating cells (silent stage). Ki-67 can label proliferating cells in any stage except those in stage G0, while it is not expressed in cells in silent stage. Ki-67 is rapidly degraded or its antigenic determinant is disappeared after mitosis. So Ki-67 is considered as a kind of objective marker reflecting proliferating activity of cells. Cyclooxygenase 2 (Cox-2) is expressed highly in cancer and it is found to be implicated in tumor progression, and its inhibition can reduce tumor growth and augment therapy.177,178 Ogunwobi179 reported that upregulation of Cox-2 could promote epithelial mesenchymal transition and carcinogenesis in vitro, and could take part in the development of HCC. Cui180 found that Cox-2 deletion significantly inhibits Ki-67 expression in the HCC xenografts. This indicates that COX-2 overexpression promotes the G1-S transition of the cell cycle in HCC xenografts which means that Cox-2 gene overexpression enhances the expression of Ki-67. Cox-2 might be taking part in the pathogenesis of HCC. Over-expression of Cox-2 has been reported to be associated with hepatocarcinogenesis. The present data revealed that the treatment with Punica granatum peel extract in HCC bearing rats resulted in significant downregulation in β-catenin expression in the liver tissue. Luteolin inhibits the translocation of βcatenin from the cytosol to nucleus in colon cancer in vitro and in vivo.181,182 Inhibition of β-catenin is mediated by modulating the expression of phosphorylated glycogen synthase kinase 3β (p-GSK3β). Also, luteolin has been shown to inhibit the expression of cyclin D1, a downstream target of Wnt/β-catenin pathway.181 Moreover, the expression of β-catenin was decreased significantly in HT29 and in HCT 116 cells treated with ellagic acid, however a marked increase in the expression of p-β-catenin levels was observed. The observed decrease in β-catenin levels and the increase in p-βcatenin expression suggests that ellagic acid effectively inactivated β-catenin and promoted its degradation and 183 thereby modulating Wnt/ β-catenin signalling cascade. Furthermore, it has been suggested that β-catenin/Tcf4 transcriptional activity was inhibited upon treatment with EGCG in HEK293 cells induced with β-catenin/Tcf4.184 Wnt/β-catenin signaling was found to be inhibited by EGCG in breast cancer cells. Treatment with EGCG resulted in upregulation of high mobility group box (HMG-box containing protein-1), an important antagonist of Wnt signaling.185,186 EGCG reduced both proliferation and invasiveness of breast cancer cells through induction of HMG-box containing protein 1 and the subsequent downregulation of Wnt/β-catenin signaling. In light of the present data, treatment with Punica granatum peel extract in HCC bearing rats led to significant downregulation in survivin expression in the 187 liver tissue. Li reported that naringin which is a one of the flavonoid compounds present in Punica granatum extract71 induced cell apoptosis and G1 cycle arrest by International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 135 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 modulation of p21 and survivin, through inactivating βcatenin signaling pathway. β-catenin pathway has been confirmed to be an essential regulator for the expression 188 of survivin. As Zhu reported that survivin expression is inhibited by down reguating β-catenin pathway. Moreover, Siegelin189 demonstrated that some flavonoids such as kaempferol present in the Punica granatum extract exerted antitumor activity through inhibiting survivin. In line with this evidence, Jeong190 stated that kaempferol could induce apoptosis through inhibition of survivin expression. More in details, it has been demonstrated that kaempferol mediated downregulation of phosphorylated Akt, thereby further reducing survivin protein level. The blockage of the serine/threonine kinase Akt activity by kaempferol is important for inhibition of survivin because active phosphorylated Akt enhances the stability of survivin.189 The observed downregulation in the expression of Ki-67 in liver tissue due to treatment of HCC bearing rats with Punica granatum peel extract might be attributed to the antiproliferative effect exerted by kaempferol. Ackland191 reported that the reduction in cell proliferation is associated with the decreased expression of nuclear proliferation antigen Ki-67. Jeong190 demonstrated that kaempferol caused a rapid inhibition of extracellular signal-regulated kinase (ERK) phosphorylation which upon activation contributes to the proliferative responses in cells and are considered to be an essential common element of mitogenic signaling. Photomicrographs of liver tissue sections of rats in NDEA administered group (HCC group) showed anaplastic activity of hepatocytes characterized by polarity, pleomorphism and hyperchromachtic nuclei. In addition, the ratio between the nucleus and cytoplasm was 1:1 with the presence of acinar glandular structure formation. Scherer and Emmelot192 stated that in NDEA administered rats, carcinomas are preceded by the development of socalled altered foci and hyperplastic nodules. Also, Burr89 reported that there was necroinflammation with perivenular inflammatory cells and macrophages in the 193 liver tissue after NDEA administration. Gupta found that NDEA administration caused vacuolization, loss of normal hepatocellular architecture and presence of pycnotic nuclei. These findings might be attributed to that NDEA is primarily metabolized in the liver and the reactive generated metabolites are known to damage hepatocytes. NDEA is an N-nitroso alkyl compound described as an effective hepatotoxin and hepatotoxic in experimental animals, producing toxicity after repeated 10 administration. Microscopic investigation of liver tissue section of rat bearing HCC and treated with high dose of Punica granatum peel extract showed focal regenerative reaction surrounding the focal necrosed hepatocytes. ISSN 0976 – 044X Also, the portal area showed inflammatory cells infiltration and seve dilatation in the portal vein. Meanwhile hepatic parenchyma showed focal degeneration in the hepatocytes. These effects might be related to that Punica granatum extract exhibited the antitumor activity, with consequent amelioration of the carcinogenic impact of NDEA. Punica granatum has been extensively studied for its antitumor property.194 This could be attributed to its high polyphenolic contents 40 including ellagic acid and ellagitannins. Microscopic investigation of liver tissue section of rat bearing HCC and treated with low dose of Punica granatum peel extract showed proliferated cystic bile ducts, associated with regenerative reaction surrounding the nodular necrosis and degenerated hepatocytes. In addition, multiple newly formed bile ductules with fibrosis and inflammatory cells infiltration in between were observed. These findings indicate the ability of this dose of Punica granatum peel extract to promote liver regeneration after counteracting the damaging impact of the carcinogenic agent. The present results revealed that the treatment with doxorubicin (Dox) in HCC bearing rats caused marked increase in serum ALT, AST and ALP activity. Dox has a metabolic activity that causes generation of free radicals and induction of oxidative stress leading to liver tissue injury.195 Hence, Dox causes an imbalance between free oxygen radicals (ROS) and antioxidants enzymes resulting in liver damage,196,197 as indicated by the increased serum indices of liver function including ALT, AST and ALP. The elevated serum levels of liver enzymes as indicators, for hepatocellular damage has been previously reported in Dox-induced hepatoxicity model.198,199 The current data revealed that the treatment with Dox in HCC bearing rats resulted in marked decrease in AFP, CEA and GPC-3 serum levels. This finding could be attributed to the powerful apoptotic property of Dox. Dox induces 200-202 apoptosis via p53/Fas/Caspase-8 system. This drug stimulates transcriptional activity of the proapoptotic molecule p53, which induces expression of the death ligand (Fas L) leading to activation of the Fas receptor. Activated Fas receptor binds, via its cytosolic domain, to adapter proteins such as the Fas-associated death domain (FADD) and the apoptosis initiator, caspase-8.203 Activated caspase-8 cleaves Bid,204,205 producing truncated Bid, which interacts with mitochondria to cause the release of cytochrome c. The latter activates caspase9, transmitting apoptotic signals to the nucleus through downstream targets such as caspase-3 and endonucleases, ultimately causing DNA fragmentation 203 and cell death. The present findings revealed that the treatment with Dox in HCC bearing rats elicited significant decrease in International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 136 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 serum VEGF. This result could be due to the antiangiogenic activity of Dox. More in details, Lee206 showed that Dox inhibited HIF-1 transcriptional activity by blocking the binding of hypoxiainduced HIF-1 to DNA. HIF-1a knockdown or inhibition increases the sensitivity of hypoxic tumor cells to Dox.207,208 Because HIF-1 is a master regulator for many aspects in cancer biology, the inhibition of HIF-1a leads to the disruption of multiple important mechanisms for tumor cell survival, angiogenesis, and progression. Although the main toxic effects of Dox on hepatocytes include: arrest cell cycle of hepatocytes,209 oxidative stress and disruption of electron transport, generation of free radicals, and activation of NF-kB,210 doxorubicin causes cancer cells death with direct toxic effects to living cells resulting in the release of doxorubicin-DNA complex into the intercellular space. This process prevents the release of the cytokines and growth factors including hepatocyte growth factor (HGF). This explains the reduction of serum HGF upon treatment of HCC bearing rats with Dox in the current work. In light of the present data, the treatment with Dox in HCC bearing rats experienced insignificant change in serum EGF. Dox is known to produce ROS via oneelectron reduction to the corresponding semiquinone free radicals that then react rapidly with oxygen to generate superoxide radical anions.211 Thus, Dox treatment results in accumulation of ROS in HCT-116 and in various cell lines like H4IIE hepatoma cells.212 Overproduction of ROS results in Akt phosphorylation105 and by this way, Dox treatment could induce Akt phosphorylation with consequent activation of EGFR pathway.105 The present findings revealed that the treatment with Dox in HCC bearing rats caused remarkable upregulation in the expression level of ING-3 gene in the liver tissue. Deficiency of Fas expression is one of the mechanisms involved in the immune evasion by tumors. Several antitumor drugs, such as DOX causes apoptosis via Fas213 mediated activation of caspase-8 in vitro. However, the significance of Fas expression in vivo is still unclear. Dox has been found to be able to induce activation of caspase-8. The role of caspase-8 appeared to be restricted to apoptosis mediated by death receptors, such as CD95, TNF-R1, and the TRAIL receptors. But, it could be postulated that Dox induces apoptosis through molecular expression of ING-3 gene and in turn activation of caspase-8.214-216 The current results showed that the treatment with Dox in HCC bearing rats resulted in observable downregulation in Foxp-1 gene expression level in the liver tissue. Suggesting that DNA methyltransferase-1 (DNMT1) is one of the targets for Dox-induced apoptosis in cancer cells, it has been proposed that the expression levels of DNMT1 in tumor cells might be important criteria that should be taken into account to evaluate the ISSN 0976 – 044X 217 selective cytotoxicity of the drug and to determine the optimal dose regimen. Therefore, the interaction between DNMT1 and Dox might be represented on alternative mechanism for induction of apoptosis in 218 cancer cell by Dox. The present data revealed that the treatment with Dox in HCC bearing rats induced marked downregulation in βcatenin, survivin and Ki-67 expression in liver the tissue. These findings might be attributed to the antiproliferative 219 and apoptotic effects exerted by Dox. There are two proposed mechanisms by which Dox could act in cancer cells, (i) intercalation into DNA and disruption of topoisomerase-II-mediated DNA repair and (ii) generation of free radicals and their damage to cellular membranes, DNA and proteins.220 221 Denard reported that Dox inhibited proliferation through stimulating de novo synthesis of ceramide, which in turn activates CREB3L1, a transcription factor synthesized as a membrane bound precursor. Dox stimulated proteolytic cleavage of CREB3L1 by site-1 protease and site-2 protease, allowing the NH2 terminal domain to enter the nucleus. Within the nucleus, it could activate transcription of genes encoding inhibitors of the cell cycle including P21. Also, the apoptotic effect of Dox is linked to the redox activation of Dox by endothelial nitric oxide synthase (eNOS).222 These mechanisms might be contributed in eliciting the observed downregulation of the proliferative and antiapoptotic markers as a consequence of Dox treatment in HCC bearing rats. Histological investigation of liver tissue section of rats bearing HCC showed inflammatory cells forming granulomatous lesions and periportal fibrosis after Dox administration. Dox has been shown to induce accumulation of inflammatory cells.223 Moreover, it has been reported that Dox is able to cause accumulation of collagen fibers mainly in the portal area, manifested as an early sign of fibrosis.224 Furthermore, microscopic examination of liver tissue section of rats bearing HCC and treated with Dox revealed that many hepatocytes showed karyomegaly and pyknotic nuclei indicating apoptosis. Apoptosis is a common feature of hepatotoxicity induced by many anti-carcinogenic drugs. It may precede necrosis or it may occur concurrently with necrosis.225 The influence of Dox on hepatocellular carcinoma comes from its interference with the synthesis of macromolecules, covalent DNA binding and DNA crosslinking, inhibition of topoisomerase II, arresting of tumor cell cycle progression in G2 phase, induction of apoptosis and generation of reactive oxygen radicals.226 In conclusion, the present findings shed light on the underlying biochemical and molecular mechanisms in favour of N-nitrosodiethylamine induce hepatocellular carcinoma. International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 137 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 Moreover, the present study provided new evidences about the potent anticancer activity of Punica granatum peel methanolic extract in regression of hepatocellular carcinoma in rats. This effect could be ascribed to the antiangiogenic potential, apoptotic efficacy and antiproliferative capacity of the active phytochemicals in this extract. ISSN 0976 – 044X 16. Ajaikumar KB, Asheef M, Babu BH, Padikkala J, The inhibition of gastric mucosal injury by Punica granatum. (pomegranate) methanolic extract, J. Ethnopharmacol, 96, 2005, 171-176. 17. Adhami VM, Mukhtar H, Anti-oxidants from green tea and pomegranate for chemoprevention of prostate cancer, Mol. Biotechnol, 37, 2007, 52-57. 18. Abdel Moneim AE, Antioxidant activities of Punica granatum (pomegranate) peel extract on brain of rats, J. Med. Plants Res, 6, 2012, 195-199. These pathways are considered the most important targets in the therapeutic intervention against hepatocellular carcinoma. 19. Rahimi HR, Arastoo M, Ostad SN, A comprehensive review of Punica granatum (Pomegranate) properties in toxicological, pharmacological, cellular and molecular biology researches, Iran. J. Pharm. Res, 11, 2012, 385-400. REFERENCES 20. Artik N, Ceremroglu B, Murakami H, Mori T, Determination of phenolic compounds in pomegranate juice by using HPLC, Fruit Processing, 8, 1998, 492–499. 1. 2. 3. 4. Sapisochin G, Charco R, Salvage liver transplantation for HCC: an old story without consensus? J Transplant Technol Res, 1, 2011, 06. Wang Y, Shen Y, Unresectable hepatocellular carcinoma treated with transarterial chemoembolization: clinical data from a single teaching hospital, Int J Clin Exp Med, 6(5), 2013, 367-371. Zhang XL, Yu H, Xiong YY, Ma ST, Zhao L, She SF, Resveratrol downregulates myosin light chain kinase, induces apoptosis and inhibits diethylnitrosamine-induced liver tumorigenesis in rats, Int J Mol Sci, 14, 2013, 1940-1951. Harring TR, Kuten DA, Nguyen NT, Goss JA, O’Mahony CA, Orthotopic liver transplantation in patients with mixed hepatocellular carcinoma- cholangiocarcinoma, J Transplant Technol Res, 1, 2011, 104. 5. Aleem E, Elshayeb A, Elhabachi N, Mansour AR, Gowily A, Hela A, Serum IGFBP-3 is a more effective predictor than IGF-1 and IGF-2 for the development of hepatocellular carcinoma in patients with chronic HCV infection, Oncol Lett, 3(3), 2012, 704-712. 6. El-Zayadi AR, Badran HM, Barakat EM, Attia MD, Shawky S, Mohamed MK, Selim O, Saeid A, Hepatocellular carcinoma in Egypt: a single center study over a decade, World J Gastroenterol, 11(33), 2005, 5193-5198. 7. 8. 9. Yates SC, Hafez M, Beld M, Lukashov VV, Hassan Z, Carboni G, Khaled H, McMorrow M, Attia M, Goudsmit J, Hepatocellular carcinoma in Egyptians with and without a history of hepatitis B virus infection: association with hepatitis C virus (HCV) infection but not with (HCV) RNA level, Am J Trop Med Hyg, 60(4), 1999, 714-720. Lehman EM, Soliman AS, Ismail K, Hablas A, Seifeldin IA, Ramadan M, El-Hamzawy H, Shoushtari CS, Wilson ML, Patterns of hepatocellular carcinoma incidence in Egypt from a populationbased Cancer registry, Hepatol Res, 38(5), 2008, 465-473. Farazi PA, DePinho RA, Hepatocellular carcinoma pathogenesis: from genes to environment, Nat Rev Cancer, 6(9), 2006, 674-687. 10. Jose K, Kuttan R, Bhattacharya K, Effect of Emblica officinalis extract on hepatocarcinogenesis and carcinogen metabolism, Journal of clinical biochemistry and nutrition, 25, 1998, 31–39. 11. Liao DJ, Blanck A, Eneroth P, Gustafsson JA, Hällström IP, Diethylnitrosamine causes pituitary damage, disturbs hormone levels, and reduces sexual dimorphism of certain liver functions in the rat, Environ Health Perspect, 109(9), 2001, 943–947. 12. Verna L, Whysner J, Williams GM, N-Nitrodiethylamine mechanistic data and risk assessment: bioactivation, DNA-adduct formation, mutagenicity, and tumor initiation, Pharmacol Ther, 71, 1996, 57-81. 13. Genoux E, Nicolle E, Boumendjel A, Flavonoids as anticancer agents: recent progress and state of the art? Current Organic Chemistry, 15, 2011, 2608-2615. 14. Adhami VM, Khan N, Mukhtar H, Cancer chemoprevention by pomegranate: laboratory and clinical evidence, Nutrition and Cancer, 61(6), 2009, 811-815. 15. Longtin R, The pomegranate: nature’s power fruit? J. National Cancer Inst, 95, 2003, 346-348. 21. Amakura Y, Okada M, Tsuji S, Tonogai Y, Determination of phenolic acids in fruit juices by isocratic column liquid chromatography, Journal of Chromatography A, 891, 2000, 183–188. 22. El-Toumy SA, Rauwald H, Two ellagitannins from Punica granatun heartwood, Phytochemistry, 61, 2002, 971-974. 23. Solt D, Farber E, New principle for the analysis of chemical carcinogenesis, Nature, 263, 1976, 701–703. 24. Karimov KHY, Inoyatova FKH, Mukhamedova MT, Changes in some indices of the synthesis of nitric oxide during the early stages of hepatocarcinogenesis, Exp Toxicol Pathol, 55(1), 2003, 17–19. 25. Barnes JM, Paget GE, Mechanisms of toxic action, Prog Med Chem, 4, 1965, 18-38. 26. Reitman S, Frankel S, A colorimetric method for the determination of serum glutamic oxalacetic and glutamic pyruvic transaminases, Am J Clin Path, 28, 1957, 56-63. 27. Bowers GN, Mc Comb RB, A continuous spectrophotometric method for measuring the activity of serum alkaline phosphatase, Clin. Chem, 12, 1966, 70-89. 28. Shu W, Lu MM, Zhang Y, Tucker PW, Zhou D, Morrisey EE, Foxp2 and Foxp1 cooperatively regulate lung and esophagus development, Development, 134(10), 2007, 1991-2000. 29. Eshak MG, Ghaly IS, Khalil WKB, Farag IM, Ghanem KZ, Genetic alterations induced by toxic effect of thermally oxidized oil and protective role of tomatoes and carrots in mice, Journal of American Science, 6(4), 2010, 175-188. 30. Bio-Rad Laboratories Inc, Real-time PCR applications guide, Bulletin, 5279, 2006, 101. 31. Bancroft JD, Gamble M, Theory and practice of histological th techniques. 6 Ed, Churchill Livingstone-Elsevier, 2008, 433-469. 32. Drury G, Wallington A, Carleton’s histological techniques, Oxford University Press, 1980, 403-406. 33. Armitage P, Berry G, Comparison of several groups. In: statistical nd method in medical research, 2 ed., Oxford, Block well significant publication, 1987, 186-213. 34. Ramakrishnan G, Jagan S, Kamaraj S, Anandakumar P, Devaki T, Silymarin attenuated mast cell recruitment thereby decreased the expressions of matrix metalloproteinases-2 and 9 in rat liver carcinogenesis, Invest New Drugs, 27, 2009, 233–240. 35. Willimsky G, Schmidt K, Loddenkemper C, Gellermann J, Blankenstein T, Virus-induced hepatocellular carcinomas cause antigen-specific local tolerance, The Journal of Clinical Investigation, 123 (3), 2013, 1032-1043. 36. Pratt DS, Kaplan MM, Evaluation of abnormal liver-enzyme results in asymptomatic patients, N Engl J Med, 342, 2000, 1266–1271. 37. El Mesallamy HO, Metwally NS, Soliman MS, Ahmed KA, Abdel Moaty MM, The chemopreventive effect of Ginkgo biloba and Silybum marianum extracts on hepatocarcinogenesis in rats, Cancer Cell International, 11, 2011, 38. 38. Rao MEB, Dama GY, Evaluation of hepatoprotective activity of Punica granatum leaves on carbon tetrachloride induced International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 138 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 hepatotoxicity in rat, International Journal of Universal Pharmacy and Life Sciences, 1(2), 2011, 23-36. 39. Chidambara-Murthy KN, Jayaprakasha GK, Singh RP, Studies on antioxidant activity of pomegranate (Punica granatum) peel extract using in vivo models, J. Agric. Food Chem, 50, 2002, 47914795. 40. Seeram, NP, Adams LS, Henning SM, Niu Y, Zhang Y, Nair MG, Heber D, In vitro antiproliferative, apoptotic and antioxidant activities of punicalagin, ellagic acid and a total pomegranate tannin extract are enhanced in combination with other polyphenols as found in pomegranate juice, J Nutr Biochem, 16(6), 2005, 360-367. 41. Zhang Z, Hamilton SM, Stewart C, Strother A, Teel RW, Inhibition of liver microsomal cytochrome P450 activity and metabolism of the tobacco-specific nitrosamine NNK by capsaicin and ellagic acid, Anticancer Res, 13, 1993, 2341-2346. 42. Barch DH, Rundhaugen LM, Thomas PE, Kardos P, Pillay NS, Dietary ellagic acid inhibits the enzymatic activity of CYP1A1 without altering hepatic concentrations of CYP1A1 or CYP1A1 mRNA, Biochem Biophys Res Commun, 201, 1994, 1477-1482. 43. Abhijeet N, Rupali P, Effect of Punica granatum seeds on doxorubicin-induced hepatotoxicity in wistar rats, J Pharm Res, 5(9), 2012, 4625. 44. Gil MI, Tomas-Barberan FA, Hess-Pierce B, Holcroft DM, Kader AA, Antioxidant activity of pomegranate juice and its relationship with phenolic composition and processing, J. Agric. Food Chem, 48, 2000, 4581–4589. 45. Ahmed HH, El-Alfy NZ, Mahmmod FM, Yahya SMM, Regression of liver fibrosis by Punica granatum pell extract in the experimental model, World Journal of Pharmacy and Pharmaceutical Sciences, 3(5), 2014, 22-44. 46. Sadiknah NAH, EL-Maraghy SA, Ismail MF, Diethylnitrosamineinduced hepatocarcino-genesis in rats: possible chemoprevention by blueberries, African J Bioch Res, 2, 2008, 081-087. 47. Sivaramakrishnan V, Pnm S, Vrp K, Devaraj SN, Attenuation of Nnitrosodiethylamine-induced hepatocellular carcinoma by a novel flavonol-Morin, Chem Biol Interact, 171, 2008, 79-88. 48. Lazarevich NL, Review: molecular mechanisms of alphafetoprotein gene expression, Biochemistry (Moscow), 65(1), 2000, 117-133. 49. Zhou L, Liu J, Luo F, Serum tumor markers for detection of hepatocellular carcinoma, World J Gastroenterol, 12(8), 2006, 1175-1181. 50. Khien VV, Mao HV, Chinh TT, Ha PT, Bang MH, Lac BV, Hop TV, Tuan NA, Don LV, Taketa K, Satomura S, Clinical evaluation of lentil lectin-reactive alpha-fetoprotein-L3 in histology-proven hepatocellular carcinoma, Int J Biol Markers, 16, 2001, 105-111. 51. Oka H, Saito A, Ito K, Kumada T, Satomura S, Kasugai H, Osaki Y, Seki T, Kudo M, Tanaka M, Multicenter prospective analysis of newly diagnosed hepatocellular carcinoma with respect to the percentage of Lens culinaris agglutinin-reactive alpha-fetoprotein, J Gastroenterol Hepatol, 16, 2001, 1378-1383. 52. Motalleb G, Hanachi P, Fauziah O, Asmah R, Effect of Berberis vulgaris fruit extract on alpha-fetoprote gene expression and chemical carcinogen metabolizing enzymes activities in hepatocarcinogenesis rats, Iranian Journal of Cancer Prevention, 1, 2008, 33-44. 53. Premalatha B, Sachdanandam P, Effects of semecarpus anacardium nut milk extract on rat serum alpha-fetoprotein level in aflatoxin B1-mediated hepatocellular carcinoma, Fitoterapia, 70, 1999, 279-283. 54. Zimmer R, Thomas P, Mutations in the carcinoembryonic antigen gene in colorectal cancer patients: implications on liver metastasis, Cancer Res, 61, 2001, 2822–2826. 55. Macnab GM, Urbanowicz JM, Kew MC, Carcino-embryonic antigen in hepatocellular cancer, Br J Cancer, 38, 1978, 51-54. ISSN 0976 – 044X 56. Thomas P, Hems DA, The hepatic clearance of circulating native and asialo carcinoembryonic antigen by the rat, Biochem Biophys Res Comm, 67, 1975, 1205-1209. 57. Li L, Jin R, Zhang X, Lv F, Liu L, Liu D, Liu K, Li N, Chen D, Oncogenic activation of glypican-3 by c-Myc in human hepatocellular carcinoma, Hepatology, 56(4), 2012, 1380-1390. 58. Filmus J, Glypicans in growth control and cancer, Glycobiology, 11, 2001, 19-23. 59. Filmus J, Capurro M, The role of glypican-3 in the regulation of body size and cancer, Cell Cycle, 7, 2008, 2787-2790. 60. Capurro M, Wanless IR, Sherman M, Deboer G, Shi W, Miyoshi E, Filmus J, Glypican-3: a novel serum and histochemical marker for hepatocellular carcinoma, Gastroenterology, 125(1), 2003, 89–97. 61. Capurro MI, Xiang YY, Lobe C, Filmus J, Glypican-3 promotes the growth of hepatocellular carcinoma by stimulating canonical Wnt signaling, Cancer Res, 65, 2005, 6245–6254. 62. Chan KL, Guan XY, Ng IO, High-throughput tissue microarray analysis of c-myc activation in chronic liver diseases and hepatocellular carcinoma, Hum Pathol, 35, 2004, 1324-1331. 63. Suzuki M1, Sugimoto K, Tanaka J, Tameda M, Inagaki Y, Kusagawa S, Nojiri K, Beppu T, Yoneda K, Yamamoto N, Ito M, Yoneda M, Uchida K, Takase K, Shiraki K, Up-regulation of glypican-3 in human hepatocellular carcinoma, Anticancer Res, 30(12), 2010, 50555061. 64. Buettner R, Mora LB, Jove R, Activated STAT signaling in human tumors provides novel molecular targets for therapeutic intervention, Clin Cancer Res, 8(4), 2002, 945-954. 65. Fuke H, Shiraki K, Sugimoto K, Tanaka J, Beppu T, Yoneda K, Yamamoto N, Ito K, Masuya M, Takei Y, Jak inhibitor induces S phase cell-cycle arrest and augments TRAIL-induced apoptosis in human hepatocellular carcinoma cells, Biochem Biophys Res Commun, 363, 2007, 738-744. 66. Edderkaoui M, Odinokova I, Ohno I, Gukovsky I, Go VL, Pandol SJ, Gukovskaya AS, Ellagic acid induces apoptosis through inhibition of nuclear factor kappa B in pancreatic cancer cells, World J Gastroenterol, 14(23), 2008, 3672-3680. 67. Sagar SM, Yance D, Wong RK, Natural health products that inhibit angiogenesis: a potential source for investigational new agents to treat cancer-Part 1, Curr. Oncol, 13(1), 2006, 14–26. 68. Priyadarsini KI, Khopde SM, Kumar SS, Mohan H, Free radical studies of ellagic acid, a natural phenolic antioxidant, J. Agric. Food Chem, 50(7), 2002, 2200–2206. 69. Han DH, Lee MJ, Kim JH, Antioxidant and apoptosis-inducing activities of ellagic acid, Anticancer Res, 26, 2006, 3601–3606. 70. Lucioli S, Anthocyanins: mechanism of action and therapeutic efficacy, Research Signpost, 37/661(2), 2012, 27-57. 71. Wang R, Ding Y, Liu R, Xiang L, Du L, Pomegranate: constituents, bioactivities and pharmacokinetics, Fruit, Vegetable and Cereal Science and Biotechnology, Global Science Books, 4(2), 2010, 7787. 72. Aggarwal BB, Shishodia S, Molecular targets of dietary agents for prevention and therapy of cancer, Biochem Pharmacol, 71, 2006, 1397-1421. 73. Chang J, Hsu Y, Kuo P, Kuo Y, Chiang L, Lin C, Increase of Bax/Bcl-XL ratio and arrest of cell cycle by luteolin in immortalised human hepatoma cell line, Life Sci, 76, 2005, 1883-1893. 74. Selvendiran K, Koga H, Ueno T, Yoshida T, Maeyama M, TorimuraT, Yano H, Kojiro M, Sata M, Luteolin promotes degradation in signal transducer and activator of transcription 3 in human hepatoma cells: an implication for the antitumor potential of flavonoids, Cancer Res, 66(9), 2006, 4826–4834. 75. Lin Q, Lai R, Chirieac LR, Li C, Thomazy VA, Grammatikakis I, Rassidakis GZ, Zhang W, Fujio Y, Kunisada K, Hamilton SR, Amin HM, Constitutive activation of JAK3/STAT3 in colon carcinoma tumors and cell lines: inhibition of JAK3/STAT3 signaling induces apoptosis and cell cycle arrest of colon carcinoma cells, Am J Pathol, 167(4), 2005, 969–980. International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 139 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 76. Liu JG, Zhao HJ, Liu YJ, Wang XL, Effect of selenium-enriched malt on VEGF and several relevant angiogenic cytokines in diethylnitrosamine induced hepatocarcinoma rats, J Trace Elem Med Biol, 24(1), 2010, 52-57. 77. Hiramoto K, Ryuno Y, Kikugawa K, Decomposition of Nnitrosamines and concomitant release of nitric oxide by Fenton reagent under physiological conditions, Mutat Res, 520, 2002, 103–111. 78. Ueno S, Aoki D, Kubo F, Hiwatashi K, Matsushita K, Oyama T, Maruyama I, AikouT, Roxithromycin inhibits constitutive activation of nuclear factor-κB by diminishing oxidative stress in a rat model of hepatocellular carcinoma, Clin. Cancer. Res, 11, 2005, 5645– 5650. 79. Forstermann U, Closs EI, Pollock JS, Nakane M, Schwarz P, Gath I, Kleinert H, Nitric oxide synthase isozymes. characterization, purification, molecular cloning, and functions, Hypertension, 23, 1994, 1121–1131. 80. Coffey BJ, Biederman J, Geller DA, Spencer T, Park KS, Shapiro SJ, Garfield SB, The course of Tourette's disorder: a literature review, Harv Rev Psychiatry, 8(4), 2000, 192–198. 81. Weiming X, Zhi L, Marilena LMA, Ian GC, The role of nitric oxide in cancer, Cell Res, 12, 2002, 311–320. 82. Chen J, Yan Y, Li J, Ma Q, Stoner GD, Ye J, Huang C, Differential requirement of signal pathways for benzo [a] pyrene (B [a] P)induced nitric oxide synthase (iNOS) in rat esophageal epithelial cells, Carcinogenesis, 26, 2005, 1035–1043. 83. Dulak J, Jozkowicz A, Dembinska-Kiec A, Wojtowicz A, Szuba A, Cooke JP, Nitric oxide induces vascular endothelial growth factor-A synthesis by rat vascular smooth muscle cells, Arterioscler Thromb Vasc Biol, 20, 2000, 659–666. 84. Jozkowicz A, Cooke JP, Guevara I, Huk I, Funovics P, Pachinger O, Weidinger F, Dulac J, Genetic augmentation of nitric oxide synthase increases the vascular generation of VEGF. Cardiovascular Research, 51, 2001, 773-783. 85. Ferrara N, Davis-Smyth T, The biology of vascular endothelial growth factor, Endocr Rev, 18, 1997, 4–25. 86. Kimura H, Weisz A, Kurashima Y, Hashimoto K, Ogura T, D’Acquisto F, Addeo R, Makuuchi M, Esumi H, The hypoxia response element of the human vascular endothelial growth factor gene mediates transcriptional regulation by nitric oxide: control of hypoxiainducible factor-1 activity by nitric oxide, Blood, 95, 2000, 189– 196. 87. Xin X, Yang S, Ingle G, Zlot C, Rangell L, Kowalski J, Schwall R, Ferrara N, Gerritsen ME, Hepatocyte growth factor enhances vascular endothelial growth factor-induced angiogenesis in vitro and in vivo, American Journal of Pathology, 158(3), 2001, 1111– 1120. 88. Zhang YW, Su Y, Volpert OV, VandeWoude GF, Hepatocyte growth factor/scatter factor mediates angiogenesis through positive VEGF and negative thrombospondin1 regulation, Proceedings of the National Academy of Sciences of the United States of America, 100(22), 2003, 12718–12723. 89. Burr AW, Hillan KJ, Mclaughlin KE, Ferrier R, Chapman C, Mathew J, Burt AD, Hepatocyte growth factor levels in liver and serum increase during chemical hepatocarcinogenesis, Hepatol, 24(5), 1996, 1282-1287. 90. Shiota G, Okano J, Kawasaki H, Kawamoto T, Nakamura T, Serum hepatocyte growth factor levels in liver diseases: clinical implications, Hepatology, 21, 1995, 106–112. 91. Nakamura T, Sakai K, Nakamura T, Matsumoto K, Hepatocyte growth factor twenty years on: much more than a growth factor, Journal of Gastroenterology and Hepatology, 26, 2011, 188–202. 92. Shimomura T, Kondo J, Ochiai M, Naka D, Miyazawa K, Morimoto Y, Kitamura N, Activation of the zymogen of hepatocyte growth factor activator by thrombin, Journal of Biological Chemistry, 268(30), 1993, 22927–22932. 93. Appleman LJ, MET signaling pathway: a rational target for cancer therapy, Journal of Clinical Oncology, 29, 2011, 4837–4838. ISSN 0976 – 044X 94. Breuhahn K, Longerich T, Schirmacher P, Dysregulation of growth factor signaling in human hepatocellular carcinoma, Oncogene, 25, 2006, 3787–3800. 95. Korn WM, Moving toward an understanding of the metastatic process in hepatocellular carcinoma, World Journal of Gastroenterology, 7(6), 2001, 777–778. 96. Osada S, Kanematsu M, Imai H, Goshima S, Clinical significance of serum HGF and c-Met expression in tumor tissue for evaluation of properties and treatment of hepatocellular carcinoma, HepatoGastroenterology, 55(82-83), 2008, 544–549. 97. Vejchapipat P, Tangkijvanich P, Theamboonlers A, Chongsrisawat V, Chittmittrapap S, Poovorawan Y, Association between serum hepatocyte growth factor and survival in untreated hepatocellular carcinoma, J. Gastroenterol, 39, 2004, 1182–1188. 98. Yamagamim H, Moriyama M, Matsumura H, Aoki H, Shimizu T, Saito T, Kaneko M, Shioda A, Tanaka N, Arakawa Y, Serum concentrations of human hepatocyte growth factor is a useful indicator for predicting the occurrence of hepatocellular carcinomas in C-viral chronic liver diseases, Cancer, 95, 2002, 824– 834. 99. Nalesnik MA, Michalopoulos GK, Growth factor pathways in development and progression of hepatocellular carcinoma, Frontiers in Bioscience, 4, 2012, 1487–1515. 100. Broten J, Michalopoulos G, Petersen B, Cruise J, Adrenergic stimulation of hepatocyte growth factor expression, Biochemical and Biophysical Research Communications, 262(1), 1999, 76–79. 101. Okamoto T, Sanda T, Asamitsu K, NF-κB signaling and carcinogenesis, Curr. Pharm. Des, 13, 2007, 447–462. 102. Preiss D, Sattar N, Non-alcoholic fatty liver disease: an overview of prevalence, diagnosis, pathogenesis and treatment considerations, Clinical Science, 115, 2008, 141–150. 103. Semela D, Dufour JF, Angiogenesis and hepatocellular carcinoma, J Hepatol, 41, 2004, 864-880. 104. Folkman J, Fundamental concepts of the angiogenic process, Curr Mol Med, 3, 2003, 643-651. 105. Sancho P, Fabregat I, Signal transduction: NADPH oxidase NOX1 controls autocrine growth of liver tumor cells through upregulation of the epidermal growth receptor pathway, J Biol Chem, 285, 2010, 24815-24824. 106. Wang N, Wang ZY, Mo SL, Loo TY, Wang DM, Luo HB, Yang DP, Chen YL, Shen JG, Chen JP, Ellagic acid, a phenolic compound, exerts anti-angiogenesis effects via VEGFR-2 signaling pathway in breast cancer, Breast Cancer Res Treat, 134(3), 2012, 943–955. 107. Labrecque L, Lamy S, Chapus A, Mihoubi S, Durocher Y, Cass B, Bojanowski MW, Gingras D, Beliveau R, Combined inhibition of PDGF and VEGF receptors by ellagic acid, a dietary-derived phenolic compound, Carcinogenesis, 26(4), 2005, 821-826. 108. Galvano F, La Fauci L, Lazzarino G, Fogliano V, Ritieni A, Ciappellano S, Battistini NC, Tavazzi B, Galvano G, Cyanidins: metabolism and biological properties, J Nutritional Biochem, 15, 2004, 2-11. 109. Hou DX, Ose T, Lin S, Harazoro K, Imamura I, Kubo M, Uto T, Terahara N, Yoshimoto M, Fujii M, Anthocyanidins induce apoptosis in human promyelocytic leukemia cells: structureactivity relationship and mechanisms involved, Int J Oncol, 23(3), 2003, 705-712. 110. Luo H, Rankin GO, Juliano N, Jiang BH, Chen YC, Kaempferol inhibits VEGF expression and in vitro angiogenesis through a novel ERK-NFkB-cMyc- p21 pathway, Food Chemistry, 130(2), 2012, 321– 328. 111. Luo H, Rankin GO, Liu L, Daddysman MK, Jiang BH, Chen YC, Kaempferol inhibits angiogenesis and VEGF expression through both HIF dependent and independent pathways in human ovarian cancer cells, Nutrition and Cancer, 61(4), 2009, 554–563. 112. Bhatia D, Thoppil RJ, Mandal A, Samtani KA, Darvesh AS, Bishayee A, Pomegranate bioactive constituents suppress cell proliferation and induce apoptosis in an experimental model of hepatocellular carcinoma: role of Wnt/β-catenin signaling pathway, Evidence International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 140 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 Based Complementary and Alternative Medicine, 2013, 2013, 371813. 113. Zhou Q, Yan B, Hu X, Li XB, Zhang J, Fang J, Luteolin inhibits invasion of prostate cancer PC3 cells through E-cadherin, Mol Cancer Ther, 8, 2009, 1684–1691. 114. Hong Z, Cao X, Li N, Zhang Y, Lan L, Zhou Y, Pan X, Shen L, Yin Z, Luo L, Luteolin is effective in the non-small cell lung cancer model with L858R/T790M EGF receptor mutation and erlotinib resistance, Br J Pharmacol, 171(11), 2014, 2842–2853. 115. Lee WJ, Wu LF, Chen WK, Wang CJ, Tseng TH, Inhibitory effect of luteolin on hepatocyte growth factor/scatter factor-induced HepG2 cell invasion involving both MAPK/ERKs and PI3K-Akt pathways, Chem Biol Interact, 160, 2006, 123–133. 116. Huang YT, Hwang JJ, Lee PP, Ke FC, Huang JH, Huang CJ, Kandaswami C, Middleton E J, Lee MT, Effects of luteolin and quercetin, inhibitors of tyrosine kinase, on growth and metastasisassociated properties in A431 cells overexpressing epidermal growth factor receptor, Br J Pharmacol, 128, 1999, 999-1010. 117. Han DH, Denison MS, Tachibana H, Yamada K, Relationship between estrogen receptor-binding and estrogenic activities of environmental estrogens and suppression by flavonoids, Biosci Biotechnol Biochem, 66, 2009,1479–1487. 118. Yang HY, Liu HL, Tian LT, Song RP, Song X, Yin DL, Liang YJ, Qu LD, Jiang HC, Liu JR, Liu LX, Expression and prognostic value of ING3 in human primary hepatocellular carcinoma, Experimental Biology and Medicine, 237, 2012, 352–361. 119. Lu M, Chen F, Wang Q, Wang K, Pan Q, Zhang X, Downregulation of inhibitor of growth 3 is correlated with tumorigenesis and progression of hepatocellular carcinoma, Oncology Letters, 4, 2012, 47-52. 120. Chen G, Wang Y, Garate M, Zhou J, Li G, The tumor suppressor ING3 is degraded by SCF (Skp2)-mediated ubiquitin-proteasome system, Oncogene, 29(10), 2010, 1498–1508. 121. Caruso ML, Valentini AM, Overexpression of p53 in a large series of patients with hepatocellular carcinoma: a clinicopathological correlation, Anticancer Res, 19, 1999, 3853–3856. 122. Lee KC, Crowe AJ, Barton MC, p53-mediated repression of alphafetoprotein gene expression by specific DNA binding, Mol Cell Biol, 19, 1999, 1279–1288. ISSN 0976 – 044X Methylation mediated silencing of microRNA-1 gene and its role in hepatocellular carcinogenesis, Cancer Res, 68(13), 2008, 5049– 5058. 132. Li D, Yang P, Li H, Cheng P, Zhang L, Wei D, Su X, Peng J, Gao H, Tan Y, Zhao Z, Li Y, Qi Z, Rui Y, Zhang T. MicroRNA-1 inhibits proliferation of hepatocarcinoma cells by targeting endothelin-1, Life Sci, 91, 2012, 440–447. 133. Hudson RS, Yi M, Esposito D, Watkins SK, Hurwitz AA, Yfantis HG, Lee DH, Borin JF, Naslund MJ, Alexander RB, Dorsey TH, Stephens RM, Croce CM, Ambs S, MicroRNA-1 is a candidate tumor suppressor and prognostic marker in human prostate cancer, Nucleic Acids Res, 40, 2012, 3689–3703. 134. Kojima S, Chiyomaru T, Kawakami K, Yoshino H, Enokida H, Nohata N, Fuse M, Ichikawa T, Naya Y, Nakagawa M, Seki N, Tumour suppressors miR-1 and miR-133a target the oncogenic function of purine nucleoside phosphorylase (PNP) in prostate cancer, Br J Cancer, 106, 2012, 405–413. 135. Leone V, D’Angelo D, Rubio I, de Freitas PM, Federico A, Colamaio M, Pallante P, Medeiros-Neto G, Fusco A, MiR-1 is a tumor suppressor in thyroid carcinogenesis targeting CCND2, CXCR4, and SDF-1alpha, J Clin Endocrinol Metab, 96(9), 2011, 1388–1398. 136. Yoshino H, Chiyomaru T, Enokida H, Kawakami K, Tatarano S, Nishiyama K, Nohata N, Seki N, Nakagawa M, The tumoursuppressive function of miR-1 and miR-133a targeting TAGLN2 in bladder cancer, Br J Cancer, 104, 2011, 808–818. 137. Kawakami K, Enokida H, Chiyomaru T, Tatarano S, Yoshino H, Kagara I, Gotanda T, Tachiwada T, Nishiyama K, Nohata N, Seki N, Nakagawa M, The functional significance of miR-1 and miR-133a in renal cell carcinoma, Eur J Cancer, 48, 2012, 827–836. 138. Nasser MW, Datta J, Nuovo G, Kutay H, Motiwala T, Majumder S, Wang B, Suster S, Jacob ST, Ghoshal K, Down-regulation of microRNA-1 (miR-1) in lung cancer, Suppression of tumorigeneic property of lung cancer cells and their sensitization to doxurubicininduced apoptosis by miR-1, Journal of Biological Chemistry, 283, 2008, 33394–33405. 139. Yan D, Dong Xda E, Chen X, Wang L, Lu C, Wang J, Qu J, Tu L, MicroRNA-1/206 targets c-Met and inhibits rhabdomyosarcoma development, Journal of Biological Chemistry, 284, 2009, 29596– 29604. 123. Ogdene SK, Lee KC, Wernke-Dollries K, Stratton SA, Aronow B, Barton MC, The p53 targets chromatin structure alteration to repress alpha-fetoprotein gene expression, J Biol Chem, 276, 2001, 42057–42062. 140. Reid JF, Sokolova V, Zoni E, Lampis A, Pizzamiglio S, Bertan C, Zanutto S, Perrone F, Camerini T, Gallino G, Verderio P, Leo E, Pilotti S, MicroRNA profiling in colorectal cancer highlights miR-1 involvement in MET-dependent proliferation, Molecular Cancer Research, 10, 2012, 504–515. 124. Nagashima M, Shiseki M, Pedeux RM, Okamura S, KitahamaShiseki M, Miura K, Yokota J, Harris CC, A novel PHD-finger motif protein, p47ING3, modulates p53-mediated transcription, cell cycle control and apoptosis, Oncogene, 22, 2003, 343–350. 141. Narayanan BA, Geoffroy O, Willingham MC, Re GG, Nixon DW, P53/p21 (WAF1/CIP1) expression and its possible role in G1 arrest and apoptosis in ellagic acid treated cancer cells, Cancer Lett, 136, 1999, 215-221. 125. Luo J, Shah S, Riabowol K, Mains PE, The Caenorhabditis elegans ING-3 gene regulates ionizing radiation-induced germ-cell apoptosis in a p53-associated pathway, Genetics, 181, 2009, 473– 482. 142. Li TM, Chen GW, Su CC, Lin JG, Yeh CC, Cheng KC, Chung JG, Ellagic acid induced p53/p21 expression, G1 arrest and apoptosis in human bladder cancer T24 cells, Anticancer Research, 25, 2005, 971–979. 126. Bressac B, Kew M, Wands J, Ozturk M, Selective G to T mutations of p53 gene in hepatocellular carcinoma from southern Africa, Nature, 350, 1991, 429–431. 143. Larrosa M, Tomás-Barberán FA, Espín JC, The dietary hydrolysable tannin punicalagin releases ellagic acid that induces apoptosis in human colon adenocarcinoma Caco-2 cells by using the mitochondrial pathway, J Nutr Biochem, 17, 2006, 611-625. 127. Hsu IC, Metcalf RA, Sun T, Welsh JA, Wang NJ, Harris CC, Mutational hotspot in the p53 gene in human hepatocellular carcinomas, Nature, 350, 1991, 427–428. 128. Wang Y, Li G, ING3 promotes UV-induced apoptosis via Fas/caspase-8 pathway in melanoma cells, J Biol Chem, 281(17), 2006, 11887-11893. 129. Zhang Y, Zhang S, Wang X, Liu J, Yang L, He S, Chen L, Huang J, Prognostic significance of Foxp-1 as an oncogene in hepatocellular carcinoma, J Clin Pathol, 65(6), 2012, 528-533. 130. Koon HB, Ippolito GC, Banham AH, Tucker PW, Foxp-1: a potential therapeutic target in cancer, Expert Opin Ther Targets, 11, 2007, 955–965. 131. Datta J, Kutay H, Nasser MW, Nuovo GJ, Wang B, Majumder S, Liu CG, Volinia S, Croce CM, Schmittgen TD, Ghoshal K, Jacob ST, 144. Mishra S, Vinayak M, Anti-carcinogenic action of ellagic acid mediated via modulation of oxidative stress regulated genes in Dalton lymphoma bearing mice, Leukemia and Lymphoma, 52, 2011, 2155–2161. 145. Horinaka M, Yoshida T, Shiraishi T, Nakata S, Wakada M, Nakanishi R, Nishino H, Matsui H, Sakai T, Luteolin induces apoptosis via death receptor 5 upregulation in human malignant tumor cells, Oncogene, 24, 2005, 7180-7189. 146. Chen ZJ, Dai YQ, Kong SS, Song FF, Li LP, Ye JF, Wang RW, Zeng S, Zhou H, Jiang HD, Luteolin is a rare substrate of human catechol-Omethyltransferase favoring a para-methylation, Mol Nutr Food Res, 57(5), 2013, 877-885. International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 141 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 147. Lee WJ, Shim JY, Zhu BT, Mechanisms for the inhibition of DNA methyltransferases by tea catechins and bioflavonoids, Mol Pharmacol, 68(4), 2005, 1018-1030. 148. Fang M, Chen D, Yang C, Dietary polyphenols may affect DNA methylation, J Nutr, 137, 2007, 223–228. 149. Nakanishi K, Sakamoto M, Yamasaki S, Todo S, Hirohashi S, Akt phosphorylation is a risk factor for early disease recurrence and poor prognosis in hepatocellular carcinoma, Cancer, 103(2), 2005, 307–312. 150. Whittaker S, Marais R, Zhu AX, The role of signaling pathways in the development and treatment of hepatocellular carcinoma, Oncogene, 29(36), 2010, 4989–5005. 151. Lustig B, Behrens J, The Wnt signaling pathway and its role in tumor development, J Cancer Res Clin Oncol, 129, 2003, 199–221. 152. Clevers H, Wnt/beta-catenin signaling in development and disease, Cell, 127, 2006, 469–480. 153. Nelson WJ, Nusse R, Convergence of Wnt, beta-catenin, and cadherin pathways, Science, 303, 2004, 1483–1487. 154. de La Coste A, Romagnolo B, Billuart P, Renard CA, Buendia MA, Soubrane O, Fabre M, Chelly J, Beldjord C, Kahn A, Perret C, Somatic mutations of the beta-catenin gene are frequent in mouse and human hepatocellular carcinomas, Proc Natl Acad Sci USA, 95, 1998, 8847–8851. 155. Wong CM, Fan ST, Ng IO, Beta-catenin mutation and overexpression in hepatocellular carcinoma: clinicopathologic and prognostic significance, Cancer, 92, 2001, 136–145. 156. Suzuki T, Yano H, Nakashima Y, Nakashima O, Kojiro M, Betacatenin expression in hepatocellular carcinoma: a possible participation of beta-catenin in the dedifferentiation process, J Gastroenterol Hepatol, 17, 2002, 994-1000. 157. Harada N, Oshima H, Katoh M, Tamai Y, Oshima M, Taketo MM, Hepatocarcinogenesis in mice with beta-catenin and Haras gene mutations, Cancer Res, 64, 2004, 48-54. 158. Buendia MA, Genetics of hepatocellular carcinoma, Semin Cancer Biol, 10, 2000, 185-200. 159. Inagawa S, Itabashi M, Adachi S, Kawamoto T, Hori M, Shimazaki J, Yoshimi F, Fukao K, Expression and prognostic roles of β-Catenin in hepatocellular carcinoma, Clin Cancer Res, 8, 2002, 450-456. 160. Zhu H, Chen XP, Zhang WG, Luo SF, Zhang BX, Expression and significance of new inhibitor of apoptosis protein survivin in hepatocellular carcinoma, World J Gastroenterol, 11, 2005, 38553859. 161. Tamm I, Wang Y, Sausville E, Scudiero DA, Vigna N, Oltersdorf T, Reed JC, IAP-family protein survivin inhibits caspase activity and apoptosis induced by Fas (CD95), Bax, caspases, and anticancer drugs, Cancer Res, 58, 1998, 5315-5320. 162. Zaffroani N, Pennati M, Daidone MG, Survivin as a target for new anticancer interventions, J Cell Mol Med, 9, 2005, 360-372. 163. Altieri DC, Validating survivin as a cancer therapeutic target, Nat Rev Cancer, 3, 2003, 46-54. 164. Li F, Survivin study: what is the next wave? J Cell Physiol, 197, 2003, 8-29. 165. Zhang XD, Franco A, Myers K, Gray C, Nguyen T, Hersey P, Relation of TNF-related apoptosis-inducing ligand (TRAIL) receptor and FLICE inhibitory protein expression to TRAIL-induced apoptosis of melanoma, Cancer Res, 59(11), 1999, 2747-2753. 166. Yamanaka T, Shiraki K, Sugimoto K, Ito T, Fujikawa K, Ito M, Takase K, Moriyama M, Nakano T, Suzuki A, Chemotherapeutic agents augment TRAIL-induced apoptosis in human hepatocellular carcinoma cell lines, Hepatology, 32(3), 2000, 482–490. 167. Kteuz S, Siegmund D, Scheurich P, Wajant H, NF-KB inducers upregulate cFLIP, a cycloheximide-sensitive inhibitor of death receptor signaling, Mol Cell Biol, 21, 2001, 3964-3973. 168. Nesterov A, Lu X, Johnson M, Miller GJ, Ivashchenko Y, Kraft AS, Elevated AKT activity protects the prostate cancer cell line LNCaP from TRAIL-induced apoptosis, J Biol Chem, 276, 2001, 1076710774. ISSN 0976 – 044X 169. Degli-Esposti MA, Dougall WC, Smolak PJ, Waugh JY, Smith CA, Goodwin RG, The novel receptor TRAIL-R4 induces NF-KB and protects against TRAIL-mediated apoptosis, yet retains an incomplete death domain, Immunity, 7, 1997, 813-820. 170. Schneider P, Thome M, Burns K, Bodmer JL, Hofmann K, Kataoka T, Holler N, Tschopp J, TRAIL receptors 1 (DR4) and 2 (DR5) signal FADD dependent apoptosis and activate NF-KB, Immunity, 7(6), 1997, 831-836. 171. Hu WH, Johnson H, Shu HB, Tumor necrosis factor-related apoptosis-inducing ligand receptors signal NF-KB and JNK activation and apoptosis through distinct pathways, J Biol Chem, 247, 1999, 30603-30610. 172. Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS, Jr, NFKB antiapoptosis: induction of TRAFl and TRAF2 and c-IAPl and cIAP2 to suppress caspase-8 activation, Science, 281, 1998, 16801683. 173. Cui J, Dong BW, Liang P, Yu XL, Yu DJ, Effect of c-myc, Ki-67, MMP2 and VEGF expression on prognosis of hepatocellular carcinoma patients undergoing tumor resection, World J Gastroenterol, 10(10), 2004, 1533-1536. 174. Pizem J, Marolt VF, Luzar B, Cor A, Proliferative and apoptotic activity in hepatocellular carcinoma and surrounding nonneoplastic liver tissue, Pflugers Arch, 442(6), 2001, 174-176. 175. Umemura T, Kai S, Hasegawa R, Keita K, Kitamura Y, Nishikawa A, Hirose M, Prevention of dual promoting effects of pentachlorophenol, an environmental pollutant, on diethylnitrosamine induced hepato- and cholangio carcinogenesis in mice by green tea infusion, Carcinogenesis, 24(6), 2003, 11051109. 176. Yeh TS, Chen TC, Chen MF, Dedifferentiation of human hepatocellular carcinoma up-regulates telomerase and Ki-67 expression, Arch Surg, 135(11), 2000, 1334-1339. 177. Song T, Zhang X, Wang C, Wu Y, Dong J, Gao J, Cai W, Hong B, Expression of miR-143 reduces growth and migration of human bladder carcinoma cells by targeting cyclooxygenase-2, Asian Pac J Cancer Prev, 12(4), 2011, 929-933. 178. Kirane A, Toombs JE, Ostapoff KT, Carbon JG, Zaknoen S, Braunfeld JB, Schwarz RE, Burrows FJ, Brekken RA, Apricoxib, a novel inhibitor of COX-2 markedly improves standard therapy response in molecularly defined models of pancreatic cancer, Clin Cancer Res, 18(18), 2012, 5031-5042. 179. Ogunwobi OO, Liu C, Hepatocyte growth factor upregulation promotes carcinogenesis and epithelial mesenchymal transition in hepatocellular carcinoma via Akt and COX-2 pathways, Clin Exp Metastasis, 28(8), 2011, 721-731. 180. Cui W, Hu SX, Tang Z, Hu1 K, In vivo effects of cyclooxygenase-2 deletion on cellular signaling in hepatocellular carcinoma xenografts in nude mice. J. Cancer Mol, 3(2), 2007, 49-54. 181. Pandurangan AK, Dharmalingam P, Sadagopan SK, Ramar M, Munusamy A, Ganapasam S, Luteolin induces growth arrest in colon cancer cells through involvement of Wnt/β-catenin/GSK-3β signaling, J Environ Pathol Toxicol Oncol, 32(2), 2013, 131-139. 182. Ashokkumar P, Sudhandiran G, Luteolin inhibits cell proliferation during azoxymethane-induced experimental colon carcinogenesis via Wnt/β-catenin pathway, Invest New Drugs, 29, 2011, 273-284. 183. Fang Y, Zhou H, Xia JF, Lin JJ, Li RZ, Yang DQ, Xu MY, Li XY, Ellagic acid regulates Wnt/β-catenin signaling pathway and CDK8 in HCT 116 and HT 29 colon cancer cells, Bangladesh J Pharmacol, 10, 2015, 47-56. 184. Dashwood WM, Orner GA, Dashwood RH, Inhibition of betacatenin/Tcf activity by white tea, green tea, and epigallocatechin3-gallate (EGCG): minor contribution of H(2)O(2) at physiologically relevant EGCG concentrations, Biochem Biophys Res Commun, 296, 2002, 584–588. 185. Sarkar FH, Li Y, Wang Z, Kong D, Cellular signaling perturbation by natural products, Cell Signal, 21(11), 2009, 1541–1547. 186. Kim J, Zhang X, Rieger-Christ KM, Summerhayes IC, Wazer DE, Paulson KE, Yee AS, Suppression of Wnt signaling by the green tea International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 142 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 compound (-)-epigallocatechin-3-gallate (EGCG) in invasive breast cancer cells. Requirement of the transcriptional repressor HBP1, J Biol Chem, 281(16), 2006, 10865–10875. 187. Li H, Yang B, Huang J, Xiang T, Yin X, Wan J, Luo F, Zhang L, Li H, Rena G, Naringin inhibits growth potential of human triplenegative breast cancer cells by targeting β-catenin signaling pathway, Toxicology Letters, 220, 2013, 219–228. 188. Zhu H, Zhang G, Wang Y, Xu N, He S, Zhang W, Chen M, Liu M, Quan L, Bai J, Inhibition of ErbB2 by Herceptin reduces survivin expression via the ErbB2-beta-catenin/TCF4-survivin pathway in ErbB2-overexpressed breast cancer cells, Cancer Science, 101, 2010, 1156–1162. 189. Siegelin MD, Reuss DE, Habel A, Herold-Mende C, von Deimling A, The flavonoid kaempferol sensitizes human glioma cells to TRAILmediated apoptosis by proteasomal degradation of surviving, Molecular Cancer Therapeutics, 7, 2008, 3566–3574. 190. Jeong JC, Kim MS, Kim TH, Kim YK, Kaempferol induces cell death through ERK and Akt-dependent down-regulation of XIAP and survivin in human glioma cells, Neurochem Res, 34, 2009, 991– 1001. 191. Ackland ML, De Waarsenburg SV, Jones R, Synergistic antiproliferative action of the flavonols quercetin and kaempferol in cultured human cancer cell lines, In vivo, 19, 2005, 69-76. 192. Scherer E, Emmelot P, Kinetics of induction and growth of precancerous liver cell foci, and liver tumour formation by diethylnitrosamine in the rat, Eur J Cancer, 11, 1975, 689-696. 193. Gupta C, Vikram A, Tripathi DN, Ramarao P, Jena GB, Antioxidant and antimutagenic effect of quercetin against DEN induced hepatotoxicity in rat, Phytother Res, 24, 2010, 119-128. 194. Masaud IA, Rohin MAK, Baig AA, Mohamad N, Potential of therapeutic antioxidant compounds from pomegranate as anticancer agent, Journal of Chemical and Pharmaceutical Research, 6(11), 2014, 427-433. 195. Kim KH, Oudit GY, Backx PH, Erythropoietin protects against doxorubicin-induced cardiomyopathyn via a phosphatidylinositol3-kinase-dependent pathway, Journal of Pharmacology and Experimental Therapeutics, 324(1), 2008, 160–169. 196. Chen Y, Jungsuwadee P, Vore M, Butterfield DA, Clair DKS, Collateral damage in cancer chemotherapy: oxidative stress in non targeted tissues, Molecular Interventions, 7(3), 2007, 147–156. 197. Essick EE, Sam F, Oxidative stress and autophagy in cardiac disease, neurological disorders, aging and cancer, Oxidative Medicine and Cellular Longevity, 3(3), 2010, 168–177. 198. Andreadou I, Sigala F, Iliodromitis EK, Papaefthimiou M, Sigalas C, Aligiannis N, Savvari P, Gorgoulis V, Papalabros E, Kremastinos DT, Acute doxorubicin cardiotoxicity is successfully treated with the phytochemical oleuropein through suppression of oxidative and nitrosative stress, Journal of Molecular and Cellular Cardiology, 42(3), 2007, 549–558. 199. Cosan D, Basaran A, Gunes HV, Degirmenci I, Aral E, The effect of doxorubicin on rats that received toxic and carcinogenic benzo(a)pyrene, Folia Histochem Cytobiolo, 46(3), 2008, 367–372. 200. Friesen C, Herr I, Krammer PH, Debatin KM, Involvement of the CD95 (APO-1/FAS) receptor/ligand system in drug- induced apoptosis in leukemia cells, Nat Med, 2(5), 1996, 574–577. 201. Friesen C, Fulda S, Debatin KM, Deficient activation of the CD95 (APO-1/Fas) system in drug-resistant cells, Leukemia, 11, 1997, 1833–1841. 202. Friesen C, Fulda S, Debatin KM, Cytotoxic drugs and the CD95 pathway, Leukemia, 13, 1999, 1854-1858. 203. Cohen GM, Caspases: the executioners of apoptosis, Biochem J, 326, 1997, 1–16. 204. Li H, Zhu H, Xu CJ, Yuan J, Cleavage of Bid by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis, Cell, 94, 1998, 491–501. 205. Kim TH, Zhao Y, Barber MJ, Kuharsky DK, Yin XM, Bid-induced cytochrome c release is mediated by a pathway independent of ISSN 0976 – 044X mitochondrial permeability transition pore and Bax, J Biol Chem, 275, 2000, 39474–39481. 206. Lee K, Qian DZ, Rey S, Wei H, Liu JO, Semenza GL, Anthracycline chemotherapy inhibits HIF-1 transcriptional activity and tumorinduced mobilization of circulating angiogenic cells, Proc Natl Acad Sci USA, 106(7), 2009, 2353–2358. 207. Song X, Liu X, Chi W, Liu Y, Wei L, Wang X, Yu J, Hypoxia-induced resistance to cisplatin and doxorubicin in non-small cell lung cancer is inhibited by silencing of HIF-1alpha gene, Cancer Chemother Pharmacol, 58(6), 2006, 776–784. 208. Nardinocchi L, Puca R, Sacchi A, D' Orazi G, Inhibition of HIF-1 alpha activity by homeodomain-interacting protein kinase-2 correlates with sensitization of chemoresistant cells to undergo apoptosis, Mol Cancer, 8, 2009, 1. 209. Kassner N, Huse K, Martin H J, Gödtel-Armbrust U, Metzger A, Meineke I, Wojnowski L, Carbonyl reductase-1 is a predominant doxorubicin reductase in the human liver, Drug Metab Dispos, 36(10), 2008, 2113–2120. 210. Zhao X, Zhang J, Tong N, Chen Y, Luo Y, Protective effects of berberine on doxorubicin-induced hepatotoxicity in mice, Biological and Pharmaceutical Bulletin, 35(5), 2012, 796–800. 211. Navarro R, Martínez R, Busnadiego I, Ruiz-Larrea MB, Ruiz-Sanz JI, Doxorubicin-induced MAPK activation in hepatocyte cultures is independent of oxidant damage, Ann. N. Y. Acad. Sci, 1090, 2006, 408–418. 212. Chovolou Y, Wätjen W, Kampkötter A, Kahl R, Downregulation of NF-kappa B activation in a H4IIE transfectant insensitive to doxorubicin-induced apoptosis, Toxicology, 232, 2007, 89–98. 213. Nakamura T, Ueda Y, JuanY, Katsuda S, Takahashi H, Koh E, Fasmediated apoptosis in adriamycin induced cardiomyopathy in rats: in vivo study, Circulation, 102, 2000, 572–578. 214. Boldin MP, Goncharov TM, Goltsev YV, Wallach D, Involvement of MACH, a novel MORT1/FADD-interacting protease, in Fas/APO-1and TNF receptor-induced cell death, Cell, 85, 1996, 803. 215. Muzio M, Chinnaiyan AM, Kischkel FC, O’Rourke K, Shevchenko A, Ni J, Scaffidi C, Bretz JD, Zhang M, Gentz R, Mann M, Krammer PH, Peter ME, Dixit VM, FLICE, a novel FADD-homologous ICE/CED-3like protease, is recruited to the CD95 (Fas/APO-1) death-inducing signaling complex, Cell, 85(6), 1996, 817-827. 216. Pan G, Ni J, Wei YF, Yu G, Gentz R, Dixit VM, An antagonist decoy receptor and a death domain-containing receptor for TRAIL, Science, 277, 1997, 815. 217. Pratt WB, Ruddon RW, Ensminger WD, Maybaum J, The anticancer drugs, Oxford University Press, New York, 1994. 218. Yokochi T, Robertson KD, Doxorubicin inhibits DNMT1, resulting in conditional apoptosis, Mol Pharmacol, 66, 2004, 1415–1420. 219. Czeczuga-Semeniuk E, Wołczyński S, Dabrowska M, Dziecioł J, Anchim T, The effect of doxorubicin and retinoids on proliferation, necrosis and apoptosis in MCF-7 breast cancer cells, Folia Histochem Cytobiol, 42(4), 2004, 221-227. 220. Thorn CF, Oshiro C, Marsh S, Hernandez-Boussard T, McLeod H, Klein TE, Altman RB, Doxorubicin pathways: pharmacodynamics and adverse effects, Pharmacogenet Genomics, 21(7), 2011, 440– 446. 221. Denard B, Lee C, Ye J, Doxorubicin blocks proliferation of cancer cells through proteolytic activation of CREB3L1, Elife, 1, 2012, 90. 222. Kalivendi SV, Kotamraju S, Zhao H, Joseph J, Kalyanaraman B, Doxorubicin-induced apoptosis is associated with increased transcription of endothelial nitric-oxide synthase, The Journal of Biological Chemistry, 276(50), 2011, 47266 –47276. 223. Saad SY, Najja TA, Al-Rikabi AC. The preventive role of deferoxamine against acute doxorubicin-induced cardiac, renal and hepatic toxicity in rats, Pharmacol Res, 43, 2001, 211-218. 224. El-Sayyad HI, Iasmail MF, Shalaby FM, Abou-El Magd RF, Gaur RL, Fernando A, Raj MHG, Ouhtit A, Histopathological effects of cisplatin, doxorubicin and 5-flurouracil (5-Fu) on the liver of male albino rats, Journal List Int Biol Sci, 5(5), 2009, 466-473. International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 143 © Copyright pro Int. J. Pharm. Sci. Rev. Res., 32(2), May – June 2015; Article No. 22, Pages: 121-144 225. Faber E, Programmed cell death: necrosis versus apoptosis, Mod. Pathol, 7, 1994, 605-609. ISSN 0976 – 044X 226. Łubgan D, Marczak A, Walczak M, Distel L, Jóźwiak Z, Pharmacological mechanisms of doxorubicin activity (DOX) – current state of knowledge, Przegl Lek, 63, 2006, 782-788. Source of Support: Nil, Conflict of Interest: None. International Journal of Pharmaceutical Sciences Review and Research Available online at www.globalresearchonline.net © Copyright protected. Unauthorised republication, reproduction, distribution, dissemination and copying of this document in whole or in part is strictly prohibited. 144 © Copyright pro